structural and mechanical analysis of a mouse model of

197
Structural and mechanical analysis of a mouse model of massive bone allografts and the effect of systemic anabolic parathyroid hormone therapy for graft healing by David Gregory Reynolds Submitted in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy Supervised by Professors Edward M. Schwarz and Hani A. Awad Department of Biomedical Engineering The College Arts & Sciences University of Rochester Rochester, New York 2008

Upload: others

Post on 06-Dec-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

Structural and mechanical analysis of a mouse model of massive bone allografts

and the effect of systemic anabolic parathyroid hormone therapy for graft

healing

by

David Gregory Reynolds

Submitted in Partial Fulfillment

of the

Requirements for the Degree

Doctor of Philosophy

Supervised by

Professors Edward M. Schwarz and Hani A. Awad

Department of Biomedical Engineering

The College

Arts & Sciences

University of Rochester

Rochester, New York

2008

ii

Curriculum Vitae The author was born in Rochester, NY on March 9, 1981. He attended the State

University of New York at Buffalo from 1999 to 2003, and graduated in 2003 with a

Bachelor of Science degree in Mechanical Engineering. He came to the University of

Rochester in the fall of 2003 and began graduate studies in Biomedical Engineering.

He received a Dean’s Fellowship in 2003 and 2005, and received a Master of

Sciences degree from the University of Rochester in 2005. He pursued research in

structural mechanics of bone allograft healing and potential adjuvant treatments under

the direction of Professors Edward M. Schwarz and Hani A. Awad.

iii

Acknowledgements 

I would first like to thank my advisors Hani and Eddie for inspiring me to do

this work. The aspirations for the goals they set for the research were high. This

work was exciting to me technically and scientifically, but I was also largely

motivated by the belief that the clinical impact could be substantial, and that

discovering new ways of diagnosing and treating skeletal defects would influence

patient outcomes in a meaningful way. I think that was the most important lesson that

I learned about myself: that I wanted to participate in influencing patient care on a

somewhat direct role through biomedical engineering and science. I also appreciate

the challenges they presented to me which helped me to stretch as an investigator. I

always found you had an open door to talk about everything from project details and

goals to career opportunities.

I’d also like to thank my committee members for their individual

contributions to this dissertation. I must admit that it was many of your ideas that are

incorporated into my research, and this dissertation would not have been the same

without them. Thank you Ruola for your expertise in CT imaging and for creating the

equipment that will make high-resolution, low-exposure radiography for clinical

translation of these studies possible in the future. Thank you, Regis, for the big-

picture view of the need to enhance massive allografts clinically. Your oversight of

both the lab and clinical work are inspirational and impressive. Thank you, Amy, for

iv

including me as a part of your lab for Thursday morning lab meetings. The

opportunity to present to your group directly and for the exposure to other aspects of

orthopaedic biomechanics helped round out my graduate education. Also, thank you

for challenging me to look at this project from other perspectives, and for asking

critical questions. My career path was greatly influenced by the seminar of yours that

my Dad thought I would be interested in 2001. I start with that day every time

someone asks me how I chose my career path.

Thank you, Dr. Christopher Beck, for the opportunity to lend your statistical

expertise and for discussing the details of multivariate regression analyses. Drs.

Susan Bukata and Lee Kaback: Thank you for providing me with the clinical fracture

cases from your clinical research to use in showing the potential for translational

applications of this work.

I could not have accomplished much of this work without the friendship and

expertise I found around me at the Center for Musculoskeletal Research Center and

Biomedical Engineering Department. Thank you all for your collaboration and

friendship. I’ve met some of the most interesting and impressive friends. In

particular, I’d like to thank Tony Chen for his breadth of expertise in biomedical

engineering, and for really important contributions to the Matlab program that was

generated for this dissertation. Other direct contributors include Saad Shaikh,

Colleen Hock, Laura Yanoso, Owen Papuga, and Krista Scorscone.

Sources of funding include grants from the Orthopedic Research Education

Foundation, the Musculoskeletal Transplant Foundation, the Wallace H. Coulter

v

Foundation, the National Institutes of Health (AR053459, DE017096, AR054041,

AR51469, and AR48681), and research grants from DePuy, J&J Inc.

Lastly, I’d like to thank my family. Their constant patience, love and

understanding have made it possible to focus these last few months. Thank you for

helping to keep our family unit a priority – you all mean so much to me. It has been a

challenging year with the passing of our father, and your time and comfort has been

instrumental in dealing with his sudden loss. I know that he lives on in each of us,

and that his good will, hard work and joy of learning are qualities that I hope to carry

on. Andrea, thank you for your confidence in me, and for being my cheerleader

through out all of this. You’re my number one. I love you. Also, thank you for

accommodating all the time I’ve been M.I.A. to focus on writing, I couldn't have

done it without you. This thesis is dedicated to you.

vi

Abstract 

The use of bone allografts for skeletal reconstructions is commonplace

clinically, but they are known to have incomplete healing even years after

implantation, fail to develop union, and ultimately fail due to unrepaired fatigue

damage. Identifying patients at risk for bone graft failure remains an unmet clinical

need. Additionally, developing new ways of enhancing bone healing are being

devised, so there is need for quantitative evaluation of their efficacy. The goals of

this dissertation were to evaluate the specific structural qualities that contribute to the

mechanical properties of grafted bones in a critically-sized defect in the mouse femur,

to generate a novel measure of graft-to-host union, and to evaluate parathyroid

hormone (PTH), a systemic anabolic bone therapy, for its effect on bone graft

healing.

An alternative to bone allograft from a tissue bank is to harvest bone from one

site within the patient and implant it into the skeletal defect site. This live bone

transplant is known as an autograft. In the first part of this dissertation, the two

standard options of bone grafting were evaluated over time to determine what

structural and morphological differences yielded the best mechanical performance

over time. Through this study a working toolset was created which could be used to

for evaluate novel adjuvant treatments for bone graft healing. We compared

processed bone allografts from donor mice which lack any intrinsic healing capacity,

vii

with live autografts, whose live periosteum and intrinsic healing capacity make them

the gold standard of bone graft materials. Surprisingly, autografts did not produce

more bone callus, but compared to the allograft the callus was better organized,

forming a bridge over the graft entirely. Correlations of the measures of cross

sectional geometry and volume of the callus and graft helped to explain up to 44%

and 50% of the variation in torsional strength and rigidity, respectively.

We observed that allograft-to-host union was deficient in many samples in

this model, which recapitulates the complication found clinically. Therefore, in the

second stage of this dissertation we devised an imaging analysis tool to measure the

degree of graft-to-host union from the CT images and coined it the Union Ratio. The

Union Ratio significantly improved the ability to predict torsional mechanics from

CT imaging by 8 to 26%, and was particularly critical in delineating successfully

healed allografts at these time points.

In the third section of this dissertation, we then investigated an adjuvant

treatment for enhancing the host's healing capacity to allografted bones. PTH has

been used to reverse osteoporotic bone loss and has recently been found to

significantly enhance fracture healing. Systemic PTH treatment was found to almost

double the callus bone volume and the union area on the allograft and nearly doubled

the yield torque and rigidity compared to saline treated controls within 6 weeks.

Multivariate regression models combining the Union Ratio, the host-to-host bridging

and the minimum cross-sectional polar moment of inertia could explain 71 – 84% of

the variation in biomechanical rigidity and strength, respectively.

viii

Lastly, progress towards evaluating persistent non unions from clinical case

studies, and measuring the effect of PTH to consolidate these fractures was made.

Together, these results indicate that achieving a high level of union, as

measured by the Union Ratio, is an important non invasive biometric in allograft

functional strength and can be improved with systemic intermittent PTH therapy

during healing.

ix

Table of Contents  Curriculum Vitae .......................................................................................................... ii Acknowledgements ...................................................................................................... iii Abstract ........................................................................................................................ vi Table of Contents ......................................................................................................... ix List of Figures ............................................................................................................. xii List of Tables ............................................................................................................. xiv List of Abbreviations .................................................................................................. xv Chapter 1: ...................................................................................................................... 1 

1.1  Introduction .................................................................................................... 1 1.1.1  The clinical need for bone grafting ......................................................... 2 1.1.2  Materials for segmental defect reconstruction ........................................ 4 1.1.3  Bone Healing .......................................................................................... 6 1.1.4  The murine femoral bone graft model .................................................... 8 1.1.5  Complicating factors associated with bone grafting ............................. 11 

1.1.5.1  Immunological response, pathogen transmission, and graft processing ....................................................................................... 11 

1.1.5.2  Microdamage accumulation in bone grafts .................................... 12 1.1.5.3  Immunologic response to bone grafts results in delayed union ..... 14 

1.1.6  Physical modification for enhancing bone allografts ............................ 17 1.1.7  Alternatives to bone allografts .............................................................. 19 1.1.8  Bioactive Adjuvant Therapies............................................................... 20 1.1.9  Parathyroid hormone treatment ............................................................. 23 

1.2  Overview of Dissertation ............................................................................. 25 Chapter 2: .................................................................................................................... 27 

2.1  Introduction .................................................................................................. 27 2.2  Background .................................................................................................. 29 2.3  Methods ........................................................................................................ 31 

2.3.1  Surgical methods. .................................................................................. 31 2.3.2  Micro-CT imaging and analysis. ........................................................... 33 

x

2.3.3  Biomechanical testing. .......................................................................... 36 2.3.4  Comparison and correlation statistics ................................................... 40 2.3.5  Background on linear regression statistical analysis ............................ 40 

2.4  Results .......................................................................................................... 43 2.4.1  Reduction in sample number ................................................................ 43 2.4.2  µCT-based morphology and structural indices ..................................... 44 2.4.3  Biomechanical testing ........................................................................... 49 2.4.4  Correlations between micro-CT parameters and torsional properties .. 51 

2.5  Discussion .................................................................................................... 54 Chapter 3: .................................................................................................................... 62 

3.1  Introduction .................................................................................................. 62 3.1.1  Previous attempts at quantifying union................................................. 63 

3.2  Methods ........................................................................................................ 65 3.2.1  Experimental model .............................................................................. 65 3.2.2  Union Ratio Algorithm ......................................................................... 66 3.2.3  Validation with Digital phantom .......................................................... 68 3.2.4  Statistical Analysis ................................................................................ 70 

3.3  Results .......................................................................................................... 73 3.3.1  Algorithm Validation ............................................................................ 73 3.3.2  Union Ratio of Autografts and Allografts ............................................ 73 3.3.3  Correlations between Union Ratio and Torsional Properties ............... 76 

3.4  Discussion .................................................................................................... 82 3.4.1  Clinical fracture non-union case study ................................................. 86 

Chapter 4: .................................................................................................................... 89 4.1  Introduction .................................................................................................. 89 4.2  Methods ........................................................................................................ 92 

4.2.1  Experimental Model.............................................................................. 92 4.2.2  Biomechanical Study ............................................................................ 93 4.2.3  Vascularization and Histological Study ................................................ 95 

4.2.3.1  Vascular perfusion .......................................................................... 95 4.2.3.2  Histology ........................................................................................ 96 

4.2.4  Statistical Analysis ................................................................................ 96 

xi

4.3  Results .......................................................................................................... 97 4.3.1  Bone analysis from Micro-CT imaging ................................................ 97 4.3.2  Biomechanical Testing Results ........................................................... 104 4.3.3  Callus Vascularization Results ........................................................... 109 

4.4  Discussion .................................................................................................. 113 Chapter 5: .................................................................................................................. 121 

5.1  Review of Research .................................................................................... 121 5.2  Microdamage Study ................................................................................... 123 5.3  Rationale for Future Research Directions .................................................. 126 

5.3.1  Further invesitigation of Union Metrics ............................................. 126 5.3.1.1  Other Clinical Applications .......................................................... 126 5.3.1.2  How much union is needed? When is a fracture “healed”? ......... 129 5.3.1.3  An alternative measure of overall connectivity ............................ 131 5.3.1.4  Large animal models of grafting and the application of union ratio ..

..................................................................................................... 134 5.3.2  Radiation exposure and justification for clinical CT imaging. ........... 137 5.3.3  Teriparatide Therapy ........................................................................... 140 

6  References ......................................................................................................... 144 7  Appendicies....................................................................................................... 157 

7.1  Appendix A: UnionRatio & µCT Analysis MATLAB Code ................... 157 7.2  Appendix B: PTH for fracture healing Literature Review ......................... 169 

xii

List of Figures 

Figure 1.1-1: Histological imaging of mouse allografts and autografts. .................... 10 

Figure 2.1-1: Structural allograft failure after implantation. ...................................... 28 

Figure 2.3-1: Mouse intercalary allograft model. ....................................................... 33 

Figure 2.3-2: Bone volume segmentation and quantification. .................................... 35 

Figure 2.3-3: Torsion testing apparatus and setup. .................................................... 38 

Figure 2.3-4: Structural graft modes of failure during torsion testing. ....................... 40 

Figure 2.4-1: Micro-CT images of allografts and autografts healing over time. ........ 45 

Figure 2.4-2: Mineralized callus and graft volumes during femoral allograft and

autograft healing over time. .................................................................. 47 

Figure 2.4-3: Cross-sectional PMI Analysis of a representative allograft and autograft.

.............................................................................................................. 48 

Figure 2.4-4: Cross sectional Polar Moment of Inertia Data. ..................................... 49 

Figure 2.4-5: The torsional properties of femoral allografts and autografts over time.

.............................................................................................................. 50 

Figure 2.4-6: Multivariable linear regression analysis micro-CT parameters vs

mechanical outcomes. .......................................................................... 53 

Figure 2.4-7: Torsional strength and rigidity vs mode of failure. ............................... 54 

Figure 3.2-1: Illustration of the graft-to-host Union Ratio algorithm. ........................ 67 

Figure 3.2-2: Algorithm validation using a digital model. ......................................... 69 

xiii

Figure 3.3-1: Representative micro-CT and union area images. ................................ 74 

Figure 3.3-2: Multivariable linear regression analysis. .............................................. 79 

Figure 3.3-3: Multivariable linear regression analysis of allografts ........................... 81 

Figure 3.4-1: Measuring the Union Area from clinical CT data of human patients. .. 87 

Figure 4.2-1: Experimental Design ............................................................................. 93 

Figure 4.3-1: Sagittal cross sections of grafted femurs. .............................................. 99 

Figure 4.3-2: Representative BV quantification from micro-CT imaging ............... 101 

Figure 4.3-3: Representative CT and Union Ratio images ....................................... 103 

Figure 4.3-4: Linear regressions between mechanical properties and Union Ratio. 108 

Figure 4.3-5: Vascularization of callus in Saline and PTH treated animals. ............ 110 

Figure 4.3-6: Maximum intensity projections of vascular perfusion imaging. ......... 111 

Figure 4.3-7: Multivariable linear regression results. ............................................... 112 

Figure 5.2-1 Microdamage in mouse cortical bone .................................................. 125 

Figure 5.3-1: 2D Flow connectivity example. ......................................................... 133 

Figure 5.3-2: Case study: Union area of a proximal tibia allograft ........................ 135 

xiv

List of Tables  Table 2.4-1: Specimen sample size per group ............................................................ 44 

Table 2.4-2: Distribution of allograft and autograft modes of failure in torsion over

time ........................................................................................................ 51 

Table 2.4-3:Correlation coefficients and significance levels for single variable linear

regression models of micro-CT-based estimates of graft ultimate torque

and torsional rigidity .............................................................................. 52 

Table 2.5-1: Relative resolution of routine clinical CT images to micro-CT images for

cortical bone and trabecular bone. ......................................................... 61 

Table 3.3-1: UnionRatio and Host-to-Host bridging callus results ............................ 75 

Table 3.3-2: Coefficients of determination and p-values for the univariate linear

regression of non-structural and structural independent variables TUlt and

TR. ......................................................................................................... 77 

Table 4.3-1: Micro-CT imaging parameters of grafted femurs. ............................... 102 

Table 4.3-2: Micro-CT imaging parameters of intact contralateral femurs. ............. 102 

Table 4.3-3: Torsional properties of grafted and contralateral femurs in mice treated

with PTH or saline as control............................................................... 105 

Table 4.3-4: Grafted femur mode of failure after torsion testing. ............................ 106 

Table 4.3-5: Coefficients of determination (R2) for the univariate linear regression of

structural independent variables vs. mechanical properties TR, TYield and

TUlt. ....................................................................................................... 107 

xv

List of Abbreviations 

BMC: Bone Mineral Content

BMD: Bone Mineral Density

BMP: Bone Morphogenetic Protein

BV: Bone Volume

BVF: Bone Volume Fraction

DBM: Demineralized Bone Matrix

MSC: Mesenchymal Stem Cell

PBS: Phosphate buffered saline

PMI: Polar Moment of Inertia

PTH: Parathyroid Hormone

Tb.N.: Trabecular Number

TUlt: Ultimate Torque

TR: Torsional Rigidity

Tb.Sp: Trabecular Separation

Tb.Th.: Trabecular Thickness

W: Work (energy) to failure

1

Chapter 1:   Introduction to bone grafting, Background and Overview of Dissertation   1.1 INTRODUCTION

The clinical need for bone grafting is to provide a source of material to

reconstruct skeletal defects for both the immediate restoration of mechanical function,

as well as the long-term durability of that surgical reconstruction. Bone allografts,

harvested post-mortem from organ and tissue donors, are the material of choice due to

their availability, workability, and largely positive outcomes. Unfortunately allografts

have complication and failure rates that are unacceptable, especially as patient life

expectancy after grafting is improving. There is a clinical need to identify recipients

whose grafts are at risk for failure and a need to define and validate adjuvant therapy

options which improve bone graft healing and longevity. The breadth of options

available for augmenting repair is widening as refinements are made to tissue

engineered bone substitutes which are osteoconductive and osteoinductive, easily

workable and have load-bearing capacity. New bioactive components and

coengraftment with live biologics are more commonly used than ever and are

improving outcomes. Improvements in medical imaging have given rise to routine

high-resolution 3D imaging using computed-tomography and magnetic resonance

imaging. While most clinical diagnoses such as the presence of bone fracture or

2

tumors are given based on qualitative evaluations, there is an increasing ability to

perform quantitative analysis for making diagnoses. Quantitative measures can

indicate biological function, but must be validated against functional gold standards

including strength and durability. Therefore, novel quantitative analyses need to be

developed in pre-clinical animal models and cadaveric studies where other destructive

analyses can be undertaken such as mechanical testing and histology.

In this introductory chapter, the need for bone grafts and the current state of

the art of grafting procedures will be introduced. Bone graft limitations, alternatives,

supplemental treatments for bone grafting will be presented. The state of the art in

quantitative diagnostic imaging tools will be reviewed. Finally, the objective of this

dissertation will be introduced.

1.1.1 The clinical need for bone grafting

As many as 800,000 skeletal bone grafting procedures are performed yearly in

the United States (Delloye et al. 2007). Large structural bone grafts from organ/tissue

donors are used to fill critical-sized skeletal defects which are a result of procedures

such as revision arthroplasty, surgical resection of tumors, and reconstruction after

trauma, including an increasing number of injured military service members. A

critical-size defect in bone is defined as a gap that is too wide for bone callus to form

bridging between the segments on its own. Therefore, critical defects need to be

filled during surgery in order to heal fully. Much of the 800,000 skeletal grafts used

each year are smaller and are for spinal fusion, joint arthrodesis, cranio-facial and

3

dental reconstruction. Another large category within bone grafts are products that

contain demineralized bone matrix (DBM) which comes in a variety of forms. What

remains after acid demineralization of bone is the underlying collagen matrix and

many osteoinductive factors including bone morphogenetic proteins (BMP) (Reddi

1998). DBM has been used since 1961 to stimulate bone formation, especially to

treat open fractures, non-unions, and to fill bone voids (Sharrard et al. 1961).

The need for improved success in bone grafts is critical in patients who

receive them for defects iatrogenically created after resection of tumors. Every year

there are approximately 300 new cases of Ewing’s Sarcoma in the USA, 700

osteosarcomas and 550 chondrosarcomas (Damron et al. 2007). Typical care consists

of surgical resection of the tumor often in combination with chemotherapy. The 5

year patient survival rate for non-metastatic Ewing’s Sarcoma has increased from

44% in the 1970s to 68% in the 1990s (Esiashvili et al. 2008). Overall survival rate

in children is between 55 and 68% (Desandes 2007). For limb-sparing surgery in

these children, bone grafts are often used to fill the large defect created by the

removal of the tumor and generally lead to enhanced quality of life. Alternatives are

implanting megaprostheses, limb amputation and rotationplasty but each is associated

with significant limitations and discomfort.

Other populations of patients receiving allografts include those who have

experienced complicated fractures due to trauma, patients who require spinal fusions

and arthroplasties. Unfortunately there are a growing number of soldiers

experiencing traumatic war injuries. There have been over 38,000 US troops

4

wounded-in-action during the Iraq war since 2003 (Defense 2008) and 28% of

injuries involve bone fractures (Baer et al. 2004) which have a high risk for infection,

osteomyelitis and non-union. Combinations of antibiotics and bone graft materials

would improve outcomes in these relatively high-morbidity injuries (Owens et al.

2006). The majority of spinal fusions use bone autograft material to achieve success

rates of 92 – 100% (Sandhu et al. 1999). In 1996 there were 228,000 spinal fusions in

the US and the number was increasing steadily. Unfortunately, the harvest of

autograft material is associated with donor site morbidity (Seiler et al. 2000; Hartman

et al. 2002) so alternatives such as allografts and osteogenic stimulating factors are

being investigated (Albert et al. 2006). Revision arthroplasties are often challenging

due to the need to make up for a loss of a patient’s bone stock caused by osteolysis,

stress shielding, infection, necrosis or complicated fractures (Huff et al. 2007) around

their old arthroplasty, which can be especially difficult in osteoporotic patients.

These defects are reconstructed with either intact cortical bone grafts or allograft

chips impacted into the defect as a void filler. These examples of the broad use of

allograft material in a variety of patients with diverse indications suggests that the use

of bone allografts will only continue to increase, thus further research to improve

these outcomes will have a large impact.

1.1.2 Materials for segmental defect reconstruction

There are a few different materials to select from which can be used to fill

large bone voids. The gold standard for sufficient healing capacity are live bone

5

autografts which are harvested from a secondary location within the patient’s body

and transplanted as live tissue. These are intrinsically osteoinductive have high

intrinsic healing capacity. The two major sites for autograft harvest are from the iliac

crest, and the fibula. Unfortunately, there is a limited source of autograft material

available for transplanting within a patient. There is also significant chronic pain and

morbidity associated with the site of autograft harvest (Summers et al. 1989; Younger

et al. 1989). The cortico-cancellous iliac crest can be harvested in a variety of shapes

to fit a variety of reconstructive needs, or it can be harvested and morselized to be

used as an osteoinductive supplement for bone healing (Sandhu et al. 1999). The

upper two-thirds of the fibula can be used for longer structural struts. Microsurgical

techniques can also retain the vascular connections of the fibular autografts at harvest,

and reconnect them to the vasculature of the new host location, thus preventing bone

necrosis (Doi et al. 1988). The live cellular component of autografts makes them

advantageous over allografts for their ability to form union, remodel and repair over

time. Live cells within the autograft, including osteocytes, active osteoclasts and

osteoblasts, vascular tissue, and the periosteum provide a rich population of

mesenchymal and osteoblastic cells and are active sources of the intercellular signals

to mediate healing. The live periosteum itself contains such a high density of

mesenchymal stem cells that can fill critically-sized defects in a ovine model of

intercalary defect by itself (Knothe Tate et al. 2007).

Bone allografts are used in most cases because they can be matched by size,

are somewhat osteoconductive, and allow for tendon and ligament reattachment.

6

When a joint is involved, the articular surface of a bone can have relatively good

outcome for a number of years, especially when the surface it is articulating against is

intact live articular cartilage (Hornicek et al. 1998; Shasha et al. 2003). Bone

allografts illicit little immune response after they have been processed, cleaned and

sterilized by the tissue banks, but this leaves them devitalized, thus lacking the

intrinsic cellular components that would mediate repair of the tissue. Unfortunately,

allograft procedures for large segmental defects experience failure rates of 23-43%

within 5 years of implantation. The majority of these failures occur within 2 years

(Berrey et al. 1990). Within these failed allografts non-unions are a contributing

complication in 27-34%, while 24-27% fracture and 9-16% have infection (Brigman

et al. 2004; Donati et al. 2005). Bone allografts are remarkably safe from infectious

contaminants and contaminated bone material is considered “a known but uncommon

complication” because of strict and effective processing controls (CDC MMWR

2002). Bone grafts rarely are thought to be responsible for the infections that arise

after surgery, (Loty et al. 1994) and are likely due to surgical introduction of

infectious agents.

1.1.3 Bone Healing

The healing response around massive bone allografts is similar to that of

typical fracture repair. Fracture healing involves three overlapping phases: the

inflammatory stage, the reparative phase, and the remodeling phase (Cruess et al.

1975; Einhorn 1998). Fracture causes a hematoma to form from the blood vessels

7

that are ruptured, and blood clotting takes place. The damaged tissue and hematoma

release cytokines that drive infiltration by inflammatory macrophages, fibroblasts and

capillary sprouting. Fibroblasts begin to lay down new matrix upon which vascular

tissue can form while macrophages begin removing debris such as the clot and

necrotic tissue. Upon neovascularization of the tissue this is termed granulation

tissue. This inflammatory phase allows for the involvement of mesenchymal cells

which get recruited to the fracture where they can then proliferate and differentiate

into chondrocytes. These chondrocytes begin to stabilize the fracture by laying down

cartilaginous callus which begins the reparative phase. This is the soft-tissue anlage

within which mineralization will occur by cartilaginous tissue hypertrophy and

osteoid deposition which will mineralize, making woven bone, thus generating the

initial consolidation of the fracture. This mineralized callus undergoes a prolonged

remodeling phase which transforms the callus into well-organized cortical bone

(Isaksson et al. 2008). As a master regulator of osteoclast activity, fracture

remodeling is partly governed by RANK signaling; the absence or blockade of it

causes osteopetrosis at the fracture site and inhibits revascularization within the callus

(Flick et al. 2003). Remodeling is partly mechanically regulated whereby regions

exposed to stress undergo more bone formation (Forwood et al. 1995), while areas

which lack stress are resorbed (Aro et al. 1982).

8

1.1.4 The murine femoral bone graft model

In order to study bone graft healing, and to validate adjuvant therapies, a

variety of pre-clinical animal models have been developed in the rat (Stevenson et al.

1997; Lewandrowski et al. 2002), dog (Delloye et al. 1986; Pluhar et al. 2001;

Ehrhart et al. 2008), rabbit (Hopp et al. 1989), sheep (Bresler et al. 1998; Knothe Tate

et al. 2007) and mouse (Ito et al. 2004). Mouse models for studying the skeleton

offer a means of elucidating the cellular and molecular mechanisms involved. This is

largely due to the multitude of genetically modified mice which allows for targeted

exploration of molecular pathways. Although rodent bone does undergo remodeling

by bone modeling units similar to Haversian remodeling, intact mouse bone lacks full

Haversian systems. Mouse models for studying musculoskeletal functions and

disorders are pervasive and have allowed for uncovering many aspects of bone

healing (Nunamaker 1998). In order to best study bone graft healing, our laboratory

published work in 2004 describing a novel mouse model of large structural bone

grafting in mice (Tiyapatanaputi et al. 2004).

The surgical technique of the model involves making a critically-sized bone

defect using two osteotomies to resect 4mm of the mid-diaphysis of the mouse femur.

The resected bone can be immediately returned to the defect site to model a live

autograft (auto- meaning self), or replaced with dead, processed bone graft from the

same genetic background of mouse (isograft) or a dead, processed graft from another

mouse (allograft). The femoral segments are aligned and secured adjacent to each

other using a 22 gage intramedullary pin. Although there are some discrepancies

9

between this model and the clinical setting, much of what is found clinically is

recapitulated in this model. Autograft repair is somewhat similar to fracture healing

in that a combination of endochondral bone formation occurs between the segments

of bone and intramembraneous bone arises from the periosteum. In the mouse, the

entire periosteum of the segmental autograft is activated by 2 weeks and results in

new bone formation along the entire length of the graft which generates a new

cortical shell bridges from host-to-host over the graft by 4 weeks (Figure 1.1-1).

Processed allografts lack a periosteum and thus have no intramembranous bone

formation along their length. Allograft repair is limited to the endochondral bone

formation initiated at the ends of the host adjacent to the graft and forms union by

creeping callus formation. Union onto the surface of the graft may be inhibited by a

fibrotic barrier that forms early on around the graft as a foreign body reaction.

Another attribute unique to autografts is their ability to be remodeled which is absent

in allografts for at least 4 weeks. Autografts are undoubtedly damaged due to the

osteotomies, and somewhat necrotic after having been severed from the vasculature,

which would cause osteocytes and bone lining cells to activate targeted resorption of

damaged tissue.

10

2 Weeks

Autograft

Allograft

4 WeeksA B

C D

Figure 1.1-1: Histological imaging of mouse allografts and autografts.

Histologic sections from fresh femoral autografts (A, B) and processed frozen

allografts (C, D) at 2 weeks (A, C) and 4 weeks (B, D) post-grafting, and stained with

alcian blue/hematoxylin. Bone formation along the length of autografts was

increased compared to allografts due to the periosteal bone formation. Bone union

occurred in all autografts by 4 weeks. There was also significant remodeling of the

entire autograft. In contrast, the ends of the allografts reached unions via creeping

callus from the host at the graft–host junction (arrow), and an absence of allograft

remodeling at week 4. (Adapted from Tiyapatanaputi 2004)

11

1.1.5 Complicating factors associated with bone grafting

1.1.5.1 Immunological response, pathogen transmission, and graft processing

Early studies showed that the immune response to minimally processed bone

grafts can be significant and that the major contributors were the cellular components

of the marrow and periosteum (Burwell 1963). Removal of cells and debris by

processing can be achieved by physical debridement, and cleansing with chemicals

such as surfactants and alcohols (Boyce et al. 1999). To prevent graft exposure to

pathogens, this processing needs to be done aseptically which can be expensive and

time consuming, so alternatively, less stringent pre-processing techniques followed

by a form of terminal sterilization can be used to achieve pathogen inactivation.

Proven forms of terminal sterilization are gamma irradiation (Sautin 1963; Loty et al.

1990; Nguyen et al. 2007), electron beam radiation (Lekishvili et al. 2004), and

ethylene oxide gas sterilization (Cloward 1980; Kakiuchi et al. 1996). Donor

screening is the first step in ensuring a safe graft tissue supply to eliminate those with

known infections, and those with risk factors associated with contracting infections,

but still, isolated cases can slip through. Simonds identified a case in which a donor

who was not identified as an HIV carrier because their serum was HIV-1 antibody

negative, but unfortunately 7 recipients of live organ or unprocessed-tissues

contracted the virus from the transplant (Simonds et al. 1992). Fortunately, viral

inactivation for reduction of the transmission in HIV, hepatitis and other infectious

agents was shown to be highly effective in this case, as graft processing prevented

disease transmission in all 34 recipients of processed tissues.

12

Other processing such as freeze drying (lyophilization) for final storage at

room temperatures for up to 5 years is an acceptable method under standards by the

American Association of Tissue Banks (Kagan 1998), but this has been shown to

reduce biomechanical strength both immediately after processing transplantation, and

is not recovered after subcutaneous implant (Kang et al. 1995) nor in a intercalary

implantation in a feline segmental cortical defect (Nather et al. 2004) where the freeze

dried bone was also less durable. Therefore, a balance between quality of graft

processing to reduce immunogenicity and disease transmission as well as

affordability of processing and storage on one hand, and maintining osteoconduction

and the biomechanical properties of the graft on the other hand needs to be achieved

according to the type of application.

1.1.5.2 Microdamage accumulation in bone grafts

Since bone allografts are implanted as devitalized material, they are subject to

accumulation of unrepaired microdamage which weakens the material over time

(Wheeler et al. 2005). This is suspected to be a major cause of catastrophic failures.

Elevated stress concentrations at the interface of implant hardware and bone are a

source of microcrack initiation (Zioupos et al. 1995; Huja et al. 1999). They are

localized at the interface between bone and prosthetic implant hardware for internal

fixation such as stems, plates and screws. Increased microdamage accumulation is

associated with cyclic loading of endosteal implants. In a study of a screw implanted

into dog cortical bone, microdamage accumulated quickly if loading was initiated

13

early after implantation. After 12 weeks of adaptation of the live bone to the implant,

osseointegration of the bone with the implant greatly minimized microdamage

accumulation (Huja et al. 1999). Clinically, allografts also experience an increased

fracture incidence when internal fixation devices penetrate the cortex of the allograft

(Thompson et al. 1993; Thompson et al. 2000; Enneking et al. 2001). Unfortunately,

targeted remodeling of microdamage (Burr et al. 1985) is absent in bone allografts

and they are incapable of adaptation around implants, thus there is an increased risk

of microdamage accumulation and fatigue failure in bone grafts. Targeted bone

remodeling after cyclic-loading-induced microdamage is mediated by the network of

osteocytes in bone (Bentolila et al. 1998). In healthy bone subjected to cyclic loading

osteocytes near microcracraks undergo apoptotic changes including DNA

fragmentation as indicated by TUNEL staining and pyknotic nuclei are there is

associated remodeling of these areas by bone remodeling units (Verborgt et al. 2000;

Verborgt et al. 2002). Targeted remodeling is therefore absent in acellular, processed

allografts. Quite recently a study found that the neuronal network may also be

involved in localized bone changes (Sample et al. 2008).

Techniques for visualizing microdamage have been developed since 1960

(Frost 1960). The gold standard has been to stain all pores within bone en bulk using

basic fuchsin before embedding in plastic for thick-sectioned slices (~200um) (Burr

et al. 1990). After sectioning, microcracks and diffuse microdamage are identifiable

using transmitted light or epifluorescent imaging (Huja et al. 1999) and are

quantifiable with manual image analysis and stereology techniques. Additionally,

14

bulk staining protocols with calcium-chelating fluorescent dyes such as calcein have

been formulated which allow epiflourescent and 3D confocal imaging of microcracks

(O'Brien et al. 2000; Lee et al. 2003). Other tools to identify microdamage in bone

include nonlinear resonant ultrasound spectroscopy (Muller et al. 2008), or barium

sulfate precipitation contrast enhanced computed tomography (Wang et al. 2007), or

super-high resolution synchrotron radiation computed tomography (Thurner et al.

2006). However, all of these techniques require the use of ex vivo bone samples and

thus cannot yet be applied clinically. Although microdamage accumulation is

associated with bone fatigue failure, accumulation of microdamage is currently

undetectable and unpredictable in clinical allografts and thus there are no indicating

risk factors for potential fatigue failures. This shortcoming deserves further

investigation and development of additional tools for studying it non-invasively. In

the mean time, the emphasis still remains on adjuvant therapies to enhance graft

healing to compensate for weakening bone material. There is also the need to devise

non-invasive clinical outcome measures to identify other risk factors such as non-

unions, or insufficient callus formation and organization. This dissertation identifies

potential solutions to these needs.

1.1.5.3 Immunologic response to bone grafts results in delayed union

Studies investigating tissue-type matching of allografts with their recipients

have not found conclusive evidence that the effect is substantial after processing

(Ward et al. 2008). Despite the fact that bone allografts have little immunologic

15

rejection associated with them (Pelker et al. 1989), they can experience a foreign

body reaction which limits their osteointegration, which could lead to prolonged non-

union between the graft and host bone. This foreign body reaction appears in the form

of an acellular fibrous barrier around the bone graft material which isolates the graft

from the host (Enneking et al. 2001; Tiyapatanaputi et al. 2004). This likely prevents

the penetration of bone modeling units into allografts, thus impairing allograft

revitalization and revascularization. The major complicating result of this is that the

establishment of union to allografts is slow, occurring anywhere between 8 and 18

months after implantation (Enneking et al. 2001; Ward et al. 2008). A complicating

factor has also uncovered that non-unions actually have an increased number of

osteoclasts (Laird et al. 2006) in a study of sheep intercalary bone grafts. Animals

with non-unions had greater numbers of osteoclasts on the surface of the graft than

those with unions. The cells that make up the fibrotic layer around non-unions

expressed the gene for the receptor activator of NF-κB ligand RANKL in non-union

specimens. These observations led to a recommendation that osteoclastic resorption

should probably be prevented by using bisphosphonates. On the other hand,

osteoclastic resorption has been recognized as the first important step to revitalizing

bone graft tissue. The debate over whether osteoclastic resorption is beneficial or

harmful may continue until a solution is found that imposes coupled remodeling of

bone allografts in which osteoblasts follow osteoclasts in succession. Previous studies

of cortical allografts and autografts in our lab found that allografts were deficient in

the osteoclastic stimulatory factor RANKL which corresponded to fewer osteoclasts

16

on the cortical surface (Ito et al. 2004; Tiyapatanaputi et al. 2004). Replenishing the

expression of this signaling factor, in combination with VEGF for stimulating

neovascularization using recombinant adeno-associated viral (rAAV) vectors was

found to revitalize implanted dead allograft bone. Surface remodeling and a 10-fold

increase in new bone formation on the graft was found by 4 weeks (Ito et al. 2005).

The immune system's foreign body reaction to the graft likely prevents graft-

to-host union. Non-union results in instability between the graft and host and puts

more stress on internal fixation hardware and raises the risk of failure. In addition,

non-union is the strongest indicating risk factor in patients whose allografts failed due

to fracture (Berrey et al. 1990). This indicates that a major deficit in graft durability

is a lack of union. Therefore I hypothesize that establishing union is paramount for a)

redistributing load from the implant-graft junction onto the graft-host junction,

thereby relieving the stress at the screw-bone interface, and b) providing a source for

bone modeling units to begin to revitalize the acellular, necrotic graft bone. The

extent of remodeling in massive bone allografts has been found to very limited, only

affecting ~20% of the bone graft in grafts recovered 5 to 13 years after implantation.

An approximate rate of remodeling into the graft surface is only 2 – 3 mm of depth

per year (Enneking et al. 1991; Stevenson et al. 1992; Enneking et al. 2001).

Interestingly, of allografts that are retrieved due to complications ~10% of them were

due to extensive graft resorption that is not associated with repair, remodeling or

revascularization (Wheeler et al. 2005). This indicates that there is at least a subset of

17

specimens that may be identifiable by x-ray as requiring an intervention to stimulate

osteoblastic bone formation.

1.1.6 Physical modification for enhancing bone allografts

Many strategies have attempted to overcome the low osteoinductivity,

imperfect osteoconductivity, and poor rate of remodeling of bone allografts by

physical modification. Early studies with demineralized bone matrix (DBM)

demonstrated that after leaching the mineral away, the remaining matrix contains

proteins that are remarkably osteoinductive (Urist 1965). The osteoinductive

components of DBM were later purified and categorized as the family of bone

morphogenetic proteins (BMPs). Surface demineralization of bone grafts was

attempted to expose these osteogenic factors but proved to not significantly improve

bone graft fate and there were few studies after 1987 (Dubuc et al. 1967; Pike et al.

1974; Kakiuchi et al. 1987) until recently when surface demineralization was

investigated for its potential to retain therapeutic agents for local delivery onto the

graft surface (Yazici et al. 2008).

Perforations within cortical allografts have been shown to affect remodeling in

some studies. Using a 1mm drill, radial perforations were made in allografts which

were implanted into a sheep defect for 6 months. Compared to standard allografts,

perforated grafts had increased bone callus formation, especially endosteal bone, and

the porosity of the graft was greatly increased due to remodeling (Delloye et al.

2002). Lewandrowski’s study compared standard allografts with partially

18

demineralized allografts and perforated and partially demineralized allografts. They

found increasing resorption and remodeling rates with the combination of

demineralization plus perforation (Lewandrowski et al. 2001). They also showed that

after 9 months, grafted bone with longitudinal perforations enhanced bending

stiffness of partially demineralized bone grafts, but were not stiffer than standard

allografts, while those with only demineralization were also weaker (Lewandrowski

et al. 2001). These studies raised concerns that mechanical integrity was

compromised in these more porous grafts (Lewandrowski et al. 1998; Lewandrowski

et al. 2001) and they have so far been rejected as a clinical alternative (Rees et al.

2003). A finite element study showed that bone is more sensitive to transverse

perforations than longitudinal perforations under axial or diametral compression

(Santoni et al. 2007) and so an in vivo study evaluating longitudinal perforations and

low intensity pulsed ultrasound (LIPUS) was undertaken. They have recently shown

that LIPUS and partial longitudinal perforations in cortical allografts seems to

improve incorporation, and increase torsional strength and stiffness by about 100%

over standard allografts, but due to a small number of animals (n=3) statistical

significance was not achieved (Santoni et al. 2008).

Additional work has been attempted to coat bone grafts with a biocompatible

polymer foam to enhance osteoconductivity of perforated and demineralized bone

grafts. They have found that a porous poly(propylene fumarate) (PPF)-hydroxlapatite

composite foam coating on allografts improves histologic incorporation of the graft-

host interface and higher strength than the uncoated controls (Lewandrowski et al.

19

2002) while at the same time protecting graft resorption. It is not possible to know if

perforation and demineralization were necessary or effective here as the PPF coating

on standard allografts was not evaluated.

In summary, physical modifications of cortical allografts have improved over

the last few decades and are encouraging means of altering osteoconductivity and

graft resorption. However, they have not overwhelmingly shown their worth, nor has

it been shown whether the perforations and demineralization affect internal fixation

devices such as plates with screws.

1.1.7 Alternatives to bone allografts

Other alternatives to structural bone grafts are engineered bone scaffolds, and

implantable prostheses. Bone scaffolds have been under development for many years.

They are typically engineered to be implanted into a void as a biocompatible,

osteoconductive 3D lattice upon which the host’s osteogenic cells will infiltrate and

eventually generate real bone. Generating scaffolds that are highly porous allows for

vascular invasion and space for new bone formation. Composition using

biodegradable materials makes complete substitution of the synthetic material

possible. Unfortunately, scaffold materials that are sufficiently strong for the load-

bearing requirements to fill major structural defects while at the same time providing

porosity and controlled biodegradation properties for positive long-term outcomes

have yet to be successfully implemented clinically. Large endoprostheses are also

available for skeletal reconstruction specifically generated for osteochondral,

20

intercalary or other defects. These can be effective, especially for revision surgeries,

but are also associated with complications. Therefore bone allografts remain the most

effective material for large skeletal reconstructions.

1.1.8 Bioactive Adjuvant Therapies

The need for adjuvant therapies for enhancing allografts is an unmet clinical

necessity. Beyond physical modification graft modification, as discussed in Section

1.1.5, three general strategies for actively overcoming the biological limitations of

allografts are 1) delivery of bioactive signaling molecules such as BMPs, 2) co-

engraftment of stem cells, and 3) therapeutic gene delivery locally to the cells around

the graft and 4) anabolic bone factors such as parathyroid hormone can be given

locally and systemically as a bone anabolic factor. The biological signals such as

BMPs can activate and recruit the host’s osteogenic population (Okubo et al. 2000).

Stem cell engraftment is meant to replenish the missing precursor population that can

differentiate into the terminal stage cells that can generate callus (Tsuchida et al.

2003). Delivering specific genes of interest locally to the cells around the graft

allows them to be the machines of the signaling molecules. This can elevate and

sustain the level of the signal beyond direct delivery of the signal, thus potentially

having a more pronounced effect.

Pre-clinical results suggested that proteins from the BMP family of growth-

regulatory factors improve bone callus generation around allografts (Cook et al. 2000;

Pluhar et al. 2001; Jones et al. 2006; Chen et al. 2007; Fukuroku et al. 2007). Short

21

exposure times and half-life of the factor is suspected to be the dominant reason why

such high doses are required, so creating slow-release delivery methods has been

investigated (Seeherman et al. 2004). The high level of dose required to produce

effective results also makes it an expensive treatment option. Evaluating the effect of

BMP treatment on bone healing and comparing the resulting costs is critical to

establish whether it is clinically cost effective. One study by Garrison et al. evaluated

the gains in patient outcomes, which included healing time, and the number of

secondary interventions, as a result of adjuvant BMP treatment for fractures, non-

unions and spinal fusion. They found that including adjuvant BMP in treatment

compared to conservative treatment had a moderate probability of being economically

justifiable for tibial fracture non-unions, and a low probability of cost-effectiveness

for lumbar spinal fusions (Garrison et al. 2007). Even greater dosages would likely

be required to affect massive cortical allografts.

Coengraftment of live stem cells to replace those that are inherently missing in

bone allografts has also been a method of enhancing bone allografting and fracture

healing (Bruder et al. 1994; Soltan et al. 2007). Rich sources of mesenchymal stem

cells are harvested from sources such as morselized autograft, as mentioned above, or

from bone marrow. This has been clinically adopted in the form of a paste that is

made from calcium phosphate collagen and autologous bone marrow aspirate such as

Vitoss (Meadows 2002) and CopiOs (www.Zimmer.com). Another source of

supplemental stem cells could be allogeneic mesenchymal stem cells (MSCs). They

have been reported to be immunoprivileged (Niemeyer et al. 2004) but this has not

22

yet been thoroughly evaluated since they have also been shown to induce immune

response and tissue rejection (Liu et al. 2006; Nauta et al. 2006; Kotobuki et al.

2008).

Delivery of therapeutic genes to stimulate skeletal reconstruction can be

accomplished by a variety of methods. Mesenchymal stem cells from various sources

can be harvested and modified ex vivo using viral vectors or other transfection

methods such as electroporation then implanted surgically (Aslan et al. 2006; Hidaka

et al. 2006). Naked plasmid delivered in vivo using a porous biocompatible material

has been coined a gene-activated matrix (GAM) (Fang et al. 1996; Bonadio et al.

1999). Transfection efficiency of locally delivered genes can be enhanced using

ultrasound and has shown to successfully induced gene expression and bone

formation (Sheyn et al. 2008). Engineered viral vectors carrying genes have been

investigated as direct in vivo gene transducers using adenovirus (Jane et al. 2002;

Chen et al. 2003; Tsuda et al. 2003), and adeno-associated virus (AAV) (Ito et al.

2004; Koefoed et al. 2005). Recently, successful delivery of BMP-2 genes was

enhanced using AAV vectors containing self-complementary (double-stranded) DNA

as opposed to previous single-stranded AAV vectors (Gazit et al. 2008) to induce

bone formation over mouse calvarial allografts. Regulation and control of gene

therapy using activation and suppression techniques would mitigate concerns about

unregulated expression of exogenous genes (Gafni et al. 2004). Engineered bone

graft substitutes combining multiple therapies are being designed to optimize

functional outcomes (Mihelic 2004; Tan et al. 2005).

23

Although there are many implantable therapeutics being developed for

enhancing surgical shortcomings at the time of surgery, there are few which can be

employed without surgical intervention. Since there is no way of knowing a priori

which graft procedures will result in non-unions or experience fatigue failure, it may

be hard to justify the additional expense of adjuvant treatments for all patients. Also,

many of these experimental therapeutics are not yet approved by the Food and Drug

Administration (FDA) for clinical use. Therefore, an FDA approved therapy which

requires no surgical intervention was investigated to determine its effects on bone

healing around allografts. This therapy is intermittent PTH 1-34, also known as

teriparatide, and marketed under the brand name Forteo™ by Lilly Pharmaceuticals.

1.1.9 Parathyroid hormone treatment

The mechanism of action of parathyroid hormone (PTH) is incompletely

understood, and its effects are diverse and seemingly incongruous, but they have been

studied in many settings and proven to be largely effective, safe and reliable as a

treatment. The basics about PTH’s actions will prove important to explaining the

effect on bone graft healing in Chapter 4. PTH has bi-phasic effects on skeletal bone

mass depending on the duration of exposure in the blood. Continuous upregulation of

PTH stimulates osteoclastic resorption of the skeleton, while intermittent exposure to

PTH causes bone formation (Locklin et al. 2003).

Under normal conditions, parathyroid hormone is a systemic master regulator

of calcium metabolism in the skeleton. When serum calcium concentration is low,

24

parathyroid cells secrete PTH into the blood. There are multiple targets within the

body that are regulated to affect calcium in the body. The first is the indirect

activation of osteoclasts in the bone which causes release of calcium from the bone

stores into the serum. To achieve general calcium balance, the other targets of PTH

seek to retain calcium by preventing its elimination in the urine and also to absorb as

much of the calcium from ingested food as possible. These processes start at the

kidney where the reabsorption of calcium is stimulated in the distal tubules. It is here

where activation of vitamin D takes place which then targets the intestine to promote

absorption from food in the intestine via active calcium pumps. Although the

exposure to elevated levels of PTH results in osteoclastic activation, this result is

indirect. It has been shown that osteoclasts lack PTH receptors (Lee et al. 1994).

Instead, PTH binds to receptors on osteoblasts and stromal cells (Fuller et al. 1998)

which then secrete RANKL which binds to RANK on osteoclasts to stimulate

osteoclast proliferation. The effect is calcium catabolism in the bone.

Hyperparathyroidism caused by parathyroid tumors yields continuous over-secretion

of PTH causing osteoporosis due to overactive osteoclasts (Locklin et al. 2003).

Interestingly, it has also been found that PTH can have an anabolic on the

skeleton when cyclically administered. Injections of full-length PTH (which is 84

amino acids) or just the most active region of PTH (AAs 1-34) results in a temporary

elevation of PTH in the blood which peaks within 30 minutes and lasts no more than

3 hours (Deal et al. 2003). In 1982 intermittent administration of PTH was observed

to increase bone mineral density in rats (Tam et al. 1982), and in 1995 it was shown

25

that this intermittent PTH did not stimulate osteoclast activity the same way as

continuous elevation of PTH (Uzawa et al. 1995). After a number of pre-clinical and

clinical trials Forteo was approved by the FDA in 2002 as a safe and effective

treatment for osteoporosis which can prevent fragility fractures. PTH’s effect is even

more impressive in healing bone. Fracture studies since 1999 have shown increases

callus volume, bone mineral density and strength in animals treated with intermittent

PTH (Andreassen et al. 1999). The application has broadened to off-label treatment

of fragility fracture non-unions in (Bukata et al. 2009). Preliminary data shows that

patients with prolonged non-unions after fracture who receive daily Forteo treatment

will achieve fracture consolidation with success rates of 93%. Recent reports of

PTH’s efficacy for enhancing autograft-mediated spinal fusion in a rat model (Abe et

al. 2007) are also encouraging for its continued broadening of potential utility.

Therefore in chapter 4 of this dissertation an attempt to enhance bone allografts is

made with intermittent administration of PTH.

1.2 OVERVIEW OF DISSERTATION

Two major problems with bone allografting are the relatively high failure rate,

and the lack of non-invasive outcome measures for identifying complications that will

lead to graft failure. These concepts were studied in this dissertation using an

established pre-clinical model of bone grafting in the mouse femur (Tiyapatanaputi et

al. 2004). This dissertation aims to first describe critical measures of structure of

bone graft healing from micro-computed tomography imaging that were identified

26

during these studies, showing that they correlate to the actual mechanical properties

of grafted femurs. Standard metrics for bone quantification such as bone volume,

cross-sectional geometry were unable to explain more than 50% of the variability in

biomechanical strength and stiffness. Qualitative observation of these specimens

revealed that graft-to-host union was not uniformly achieved and that regardless of

whether a specimen had union or not, there were wide distributions of bone callus

volumes and cross sectional geometry. Therefore, I hypothesized that samples with

large callus but no graft-to-callus union were clouding the ability to predict strength

based on simple measures alone. Since non-unions are also a frequent major

complicating factor of allografts clinically we determined that it was necessary to

develop a novel measure of bone graft-to-host union.

Next the characterization of the effect of systemic PTH treatment on bone

graft healing both non-invasively through imaging and mechanical testing was

performed. It was determined that PTH can efficiently overcome bone grafting non-

unions. Lastly a foray is made into the direct clinical potential for measuring non-

unions in routine CT imaging.

27

Chapter 2: Evaluation of the healing patterns of allografts and autografts by micro­CT image analysis and biomechanical torsion testing    2.1 INTRODUCTION

In cases of large skeletal insufficiency, bone grafts from organ and tissue

donors are used to replace the patient’s bone. The functional outcome after bone graft

transplantation is the recovery of the load bearing capacity to support the body and

regain function of the affected limb. Bone throughout the body is put under enormous

stress during routine activities and even greater stress during traumatic accidents.

Both routine and accidental loading can cause bone to fail, causing physical injury to

the patient. After bone grafting, a patient may regain the activities and capabilities

and achieve a high quality of life. Unfortunately, the risk of bone allograft failure is

much higher than normal bone fracture, and allografts are unlikely to heal on their

own, thus requiring further medical intervention. Figure 2.1.1 is an example of such a

case.

28

Figure 2.1-1: Structural allograft failure after implantation.

A patient with osteolytic Ewing’s sarcoma in the proximal tibia was treated by

surgical resection of the tumor and the affected bone. To spare the limb from

amputation, a cadaveric allograft (A) was used to fill the defect and secured internally

to the host bone with plates and screws (B). The arrow indicates fractures in the

metaphyseal region 1 year after implantation (C). A fibular autograft was used to

revise the grafted segment to assist in graft healing and revitalization (D). This was

held together with additional plates and screws. This second construction failed and

a prosthetic total knee arthroplasty (TKA) was performed (E) for the 14 year old

patient. [Reproduced from Awad 2007 with permission by Tissue Engineering]

To date, there are few indications for allograft failure that allow clinicians to

identify patients at risk for bone graft failure. The goal of this chapter is to describe

the state of the art in non-invasive and invasive biomechanical analysis of healing

bone and to use these tools to develop a system for analysis of bone in an animal

29

model where allografts and autografts are compared. This system is used in Chapter

4 to evaluate allografts in mice treated with PTH.

In this chapter allografts and autografts in mice were evaluated at multiple

time points to uncover the natural progression of graft healing. Their structure was

imaged and analyzed using micro-CT imaging and they were finally mechanically

tested. Statistical regression analysis was performed to identify measures of structure

which are critical for explaining the variation in biomechanical strength and rigidity.

Successes and shortcomings in this approach and the results are discussed and used to

stimulate further development of techniques and therapies.

2.2 BACKGROUND

There has been extensive research in human and experimental animal models

of bone allograft healing (Friedlaender et al. 1978; Burchardt 1983; Pelker et al.

1983; Weiland et al. 1984; Pelker et al. 1987; Pelker et al. 1989; Friedlaender 1991;

Kerry et al. 1999; Stevenson 1999; Wheeler et al. 2001; Wheeler et al. 2005), yet our

understanding of the immunologic, biologic, and biomechanical mechanisms of

allograft failure remains incomplete. What are the signals that identify grafted bone

as a foreign body which leads to its envelopment by a fibrous barrier? What specific

signals are missing from the dead allografts when microdamage occurs which limit

targeted remodeling? How are the stresses throughout the graft different in grafts that

fail by fatigue different from grafts that are enduring? To what extent is graft failure

due to graft processing, surgical technique, patient healing response, and patient

30

activity levels? These questions remain to be answered, but there have been many

attempts to find solutions to the difficulties encountered with bone grafting.

Furthermore, advances in evaluating these therapeutic adjuvants to improve bone

allograft repair have been slowed by the lack of quantitative and non-invasive

imaging-based outcome measures of graft biomechanical strength. Experimental

animal studies suggest that long bone defect repair (Bonadio et al. 1999) and allograft

healing (Ito et al. 2005; Koefoed et al. 2005) can be improved using localized gene

delivery vectors among other techniques.

An important alternative to bone allografting is autografting. An autograft is a

transfer of bone material from one location in the body to another. The most common

site of large strut autografts are from the patient's fibula. An autograft is implanted as

live bone material and thus has greater healing and reparative capacity, but is limited

in size. Furthermore, putative clinical reports suggest that improved tibial or femoral

allograft repair and revitalizing can be achieved by placing a vascularized fibular

autograft inside the massive processed allograft (Manfrini et al. 2004). However,

none of these studies reported quantitative indicators of the biomechanical strength of

the allografts. Before such outcome measures can be used in clinical applications,

they would first have to be developed and validated in preclinical animal models.

Toward the development of non-invasive assessment of the biomechanical

properties of structural bone grafts in a pre-clinical animal model, this chapter

investigates statistical correlations among micro-computed tomography (micro-CT)

imaging and biomechanical torsion testing parameters in the mouse femoral graft

31

model described in Chapter 1. In this study, the 4-mm mid-diaphyseal segment is

removed and either immediately replaced as a live autograft, or a graft from one

animal is devitalized and transplanted into another mouse as an allograft. Previous

studies helped identify critical molecular and cellular differences between autograft

and allograft healing (Tiyapatanaputi et al. 2004; Ito et al. 2005; Zhang et al. 2005;

Zhang et al. 2005); however, the structural and biomechanical aspects of both

autograft and allograft healing have yet to be studied in this model. Furthermore,

while previous animal studies have correlated biomechanical properties of long bone

fracture repair with imaging derived parameters (Markel et al. 1991; den Boer et al.

1999; Blokhuis et al. 2000; Shefelbine et al. 2005), no study has investigated

quantitative correlations in long bone segmental autograft and allograft repair in

animal models or human patients. To address these issues, we tested the hypothesis

that murine femoral autografts heal with improved biomechanical properties

compared to processed allografts. We also tested the hypothesis that micro-CT

parameters of graft and callus volume and geometry correlate significantly with the

torsional properties of the murine femoral grafts.

2.3 METHODS

2.3.1 Surgical methods.

All segmental femoral autograft and allograft surgeries were performed on 8-

week-old C57BL/6 mice following protocols that were approved by the University

Committee on Animal Resources as previously described (Tiyapatanaputi et al.

32

2004). Femurs from donor mice were harvested and processed with the following

steps. First the soft tissue and periosteum were scraped from the diaphysis of the

femur with a scalpel. The ends of the femur were then cut off using a 20mm round

diamond-sintered saw with a blade thickness of 0.2mm on a Dremel Tool to access

the marrow cavity. Using a syringe with 26 gage needle the marrow cavity was

flushed using 70% ethanol. The femurs were then trimmed to 4mm in length using

the same diamond saw and a caliper for measurement. They were then bathed in 70%

ethanol for 3 hours, rinsed three times with sterile saline and frozen to -80oC for 1

week to make cleaned, processed, aseptic, devitalized bone allografts.

Animals undergoing recovery surgery were anesthetized and an incision was

made on the lateral aspect of the left hind limb. The soft tissue and muscle were

dissected using blunt techniques to expose the midshaft of the femur. A double

osteotomy was made to remove 4 millimeters of bone. The live bone that was

resected was placed back into the defect with it's periosteum intact to be used as an

autograft or replaced with a processed allograft. Finally, the graft was secured

between the two host segments using a stainless steel intramedullary pin with a

0.35mm diameter (Figure 2.3.1). The pin is inserted through the knee, the graft is slid

over the pin, then the pin is passed through the greater trocanter of the femur. The

sharp end is cut square, then bent in a tight curl at each end to protect the soft tissue

and to allow for pin extraction after harvesting the femur. Weekly x-rays were taken

to monitor progression (Faxitron X-Ray LLC, Wheeling, IL).

33

Mice receiving either live autografts or devitalized allografts were sacrificed

at 6, 9, 12, and 18 weeks after surgery (n = 6 – 14, see Table 2.4-1) mice per

treatment group per time point). Femurs were disarticulated from the hip and knee

joints and the intramedullary stainless-steel pins were removed carefully. Specimens

were moistened with saline and frozen at -20oC until they were thawed for micro-CT

imaging and subsequent biomechanical testing.

Figure 2.3-1: Mouse intercalary allograft model.

Radiograph immediately after surgery of a 4mm intercalary bone graft implanted into

the femur of a mouse and secured with an intramedullary pin. [Reproduced from

Awad 2007 with permission from Tissue Engineering]

2.3.2 Micro-CT imaging and analysis.

Micro-computed tomography (micro-CT) Micro-CT imaging of the 6, 9, and

12 week specimens was performed at high resolution (13.9 mm) using the Explore

Locus SP scanner (GE Healthcare Technologies, London, ON) at 80 kVp, 80 mA,

415 projections, 1700 ms integration time; while the 18-week specimens were

34

scanned at high resolution (10.5 mm) on the VivaCT40 micro-CT scanner (Scanco

Medical, Basserdorf, Switzerland) at 55 kVp, 145 mA, 300 ms integration time.

Quantification of bone and graft volume was performed as previously

described using MicroView software (GE Healthcare) (Koefoed et al. 2005). Briefly,

total bone volume (BVTotal) between the graft–host interfaces was quantified. Graft

bone volume (BVGraft) was determined by manually segmenting the graft from the

surrounding mineralized callus. Mineralized callus volume (BVCallus) within the span

of the graft was computed from the difference between BVTotal and BVGraft (Figure

2.3.2). To compensate for image intensity variations of the scanner, a threshold was

determined for each scan using a standardized automatic threshold-selection feature

of the GE MicroView software that utilizes the Otsu method. This determines the

threshold which maximizes the variance between the groups of pixels (Otsu 1979).

The selected threshold was consistently verified against the user’s perception of the

boundary of the mineralized bone. To correct for small variations in length, all grafts

were measured using a digital caliper (Resolution: 70.01 mm; Model CD-600PS,

Mitutoyo Corp., Japan) and bone volumes were normalized to the measured length of

each graft. A custom-written MATLAB code (The Mathworks, Natick, MA) was

developed for computing the cross-sectional polar moment of inertia (PMI) about the

area centroid on each binarized slice of the grafted region. In circular, prismatic

shafts, the PMI correlates directly with torsional rigidity and inversely with the shear

stress (Shigley et al. 2001) and was used because it combines the quantity of the

material as well as the distance that material is from the torsional axis. Briefly,

35

numerical integration of thresholded (mineralized) pixels was performed, based on

the equation · where dA is the elemental area of each mineralized

pixel, and r is the radial distance to the element dA from the cross-section centroid.

The average, minimum, and maximum PMI (PMIAve, PMIMin, and PMIMax,

respectively) were determined for each specimen, as well as the PMI averaged over

the Middle 50% of the grafted region (PMIMid Graft) and over the proximal and distal

ends of the grafted region corresponding to the remaining 50% of the graft length on

both ends (PMIGraft Ends).

A B C

Figure 2.3-2: Bone volume segmentation and quantification.

The region of interest was defined as the region being within the proximal and distal

ends of the graft. The volume of total bone within that is quantified as BVTotal (A).

The graft is then manually segmented on 2D axial cross sections and volume is

calculated as BVGraft (B). What remains after subtraction of the graft from BVTotal

is the BVCallus (C). [Reproduced from Reynolds 2007 with permission of the

American Society of Bone and Mineral Research]

36

2.3.3 Biomechanical testing.

Evaluation of the mechanical competency of the grafted femur was desired in

order to understand how the entire bone performed. Other studies investigating the

material properties of the graft distinctly from how the host interacts with the graft

have been performed by excision and cementing of the allograft material only for

testing (Nather 2004). This isolates changes in the graft material from changes in the

callus formation. In these experiments we are interested in the grafted femur as a

whole, therefore we tested the graft, the host material at the ends of the graft, and the

connectivity between the segments by gripping the most proximal and distal ends of

the host. Torsion testing was chosen because it allows for concomitant testing of both

the graft and callus material, and their degree of connectivity, and it is a loading

condition that is experienced routinely as evidenced by a high frequency of spiral-

shaped fracture (Mellick et al. 1999). There have been many studies of whole bone

and healing bone that have used torsion testing and 3 and 4-point beam bending as an

outcome measure in studying mice (Wunder et al. 1977; Camacho et al. 1995; Mikic

et al. 1995; Brodt et al. 1999). Three- and four-point-bending were not pursued in

this study due to the possibility that the bone or callus would experience both

indentation and bending, and thus accurate interpretation of the loading and

displacement would be challenging.

After micro-CT imaging, the ends of the femurs were cemented into 6.35mm2

aluminum tube holders using PMMA in a custom jig to ensure axial alignment and to

maintain a gage length of 6.37 ± 0.9 mm, allowing at least 3mm to be potted at each

37

end. Specimens were bathed in PBS at room temperature for at least 2 h after potting

to allow for rehydration of the tissue (Broz et al. 1993) and hardening of the PMMA.

Specimens were then mounted on an EnduraTec TestBenchTM system (200Nmm

torque cell; Bose Corporation, Minnetonka, MN) and tested in torsion at a rate of 1o/s

until failure (Brodt et al. 1999) (Figure 2.3.3). The torque data were plotted against

the rotational deformation (normalized by the gage length and expressed as rad/mm)

to determine the ultimate torque (TUlt) and torsional rigidity (TR).

38

Figure 2.3-3: Torsion testing apparatus and setup.

Torque Cell

0

5

10

15

20

25

30

0 0.02 0.04 0.06 0.08

Normalized Rotation (rad/mm)

Torq

ue (N

.mm

)

AutograftAllograft

Allografted and autografted femurs were tested in torsion using a servoelectric motor

rotating at 1o/sec on the right with torque transducer on the fixed side on the left (A &

B). Femurs were potted into aluminum tubing, with bone cement, to a depth of

approximately 3mm leaving a gage length of approximately 6mm was left exposed

Normal

A

C

B

D

39

between the bone cement as the tested region of interest (C). Sample allograft,

autograft and normal femur torsion testing curves are shown in D.

After testing to failure, all samples were X-rayed to assess the mode of failure

(Lewandrowski et al. 2002). Specimens were classified into three distinct failure

modes. A ‘‘Pre-union’’ mode of failure presented as if the graft, clearly recognizable

and intact, was simply pulled out from the host callus and is likely due to very weak

union at either of the graft–host interfaces. An ‘‘Early union’’ mode of failure was

characterized by a fracture which involved the graft–host interface, but also extended

into the graft and/or host, indicating that there was some degree of union. The third

mode of failure that we observed was termed ‘‘Mature union’’ in which failure

largely occurred in the graft region as a spiral fracture, which commonly occurs when

torsional loads are applied to normal bone (Figure 2.3-4).

40

A B C

Figure 2.3-4: Structural graft modes of failure during torsion testing.

X-rays were taken of the grafted femurs after destructive torsion testing to assess the

mode of failure as defined by the fracture location and morphology. Three distinct

modes of failure were identified: ‘‘Pre-union’’ failure (A) presented as if the graft was

simply pulled out from the host callus; ‘‘Early union’’ failure (B) was characterized by

a fracture which involved the graft–host interface, but also extended further into the

graft and/or host; ‘‘Mature union’’ (C) in which failure largely occurred in the graft

region. [Reproduced from Reynolds 2007 with permission of the American Society of

Bone and Mineral Research]

2.3.4 Comparison and correlation statistics

Autograft and allograft data were compared using analysis of variance and

Bonferroni post hoc multiple comparisons. For the analysis of the mode of failure, a

chi-square test was conducted to determine whether the distribution of failure modes

was different between allografts and autografts (Tamhane 2000).

2.3.5 Background on linear regression statistical analysis

In order to elucidate the effect of structure and organization had on strength

and rigidity of bone grafts, we first investigated all of the individual parameters'

41

ability to correlate with strength and rigidity. We hypothesized that analysis of the

full set of data which included allografts and autografts at 6, 9, 12 and 18 weeks

would yield an indication of the most critical factors for improving grafted bone

strength and rigidity in an unbiased way. This resulted in multiple factors which had

significant correlations, but no single parameter was able to explain more than 32% of

the variation in mechanical properties, therefore further analysis using multiple

structural measures would be required to convincingly surmise the strength of grafted

femurs from 3D medical images.

There are a variety of ways of combining multiple independent factors to

generate an equation that relates to an outcome. Here we are trying to use multiple

structural measures to indicate the mechanical strength and rigidity outcomes. We

measured 8 different structural parameters, which means that there could be over 400

linear combinations of independent variables. There are two factors to consider when

generating an optimum regression equation. The first is that the ‘independent’

variables may be correlated to each other; therefore including both of them would be

redundant in the regression. Secondly, an independent variable that is brought into a

regression equation carries with it some uncertainty about whether the value is

actually related to the outcome, so there needs to be a penalty which accounts for

including additional terms. Two accepted methods of accounting for the problem of

“over-fitting” the regression are the Adjusted R2 which applies a penalty to the

coefficient of determination which is based on the number of regressors and the

number of samples in the dataset, or the use of Mallows’ CP (Mallows 1973).

42

Mallows’ CP is an indicator of fit defined as: 2 , where SSEP is the

error sum of squares for the model fit with P regressors, S2 is the residual mean

squared of the regression which uses the full set of independent variables and N is the

sample size. Therefore, when the next best variable is added to the regression

equation, the SSEP should go down, and P increases, while S2 and N remain constant.

When adding one-too-many variables to the regression the SSEP does not sufficiently

diminish to overcome the increase in P, thus CP rises from the previous step.

Therefore, minimization of CP indicates optimum regression fit. Other complicating

considerations are that an independent variable may be correlated to the outcome

variable, but in a non-linear way, or that terms may only be significantly important to

the outcome measure as part of a term of interacting variables. For this study we

wanted to use the data that was available in the most efficient way. The following

describes the methodology that was used and the explanation of the choices that were

made to perform these studies.

Since we know that allografts and autografts have very different healing

mechanisms, we realize that a measure for allografts might correlate to strength

differently than a measure for autografts, therefore the type of graft might be an

important co-variate. An example is that BVGraft seems to diminish greatly as

autografts mature, and is actually inversely related to biomechanical strength in

autografts (Figure 2.4-2), while BVGraft is relatively stable in allografts past 9 weeks

but that there are major changes in allograft strength from 6 to 9 weeks. The reason

for autograft resorption will be described in the discussion section of this chapter. In

43

such case, BVGraft may be a significant inverse indicator of strength, while BVGraft

may have a different effect for allografted femurs. A similar argument could be made

for the time as a covariate: BVCallus at one time point may not be important if the

specimens have not formed union at both junctions, while later on, when union is

established, variations in BVCallus may correspond to variation in bone mechanics.

Therefore, the inclusion of time and graft-type will also be considered.

All analyses were performed using SAS 9.1 (SAS Institute Inc., Cary, NC)

based on a best subset selection method which minimizes Mallows’ CP statistic to

optimize the number of independent variables and interactions included in the model

(Mallows, 1973).

2.4 RESULTS

2.4.1 Reduction in sample number

Although most groups maintained a high sample number, allografts at 6 and 9

weeks, and autografts at 6 weeks experienced multiple specimen losses. The

dominant reason for specimen loss was general weakness in grafted femurs at these

time points. Specimens were harvested and dissected carefully, but occasionally after

intramedullary pin removal it was determined that these specimens were not intact

and there was no way of distinguishing specimens that failed during harvesting from

those that had not yet attained consolidation (union) of the graft-host junction. The

rarer cause of specimen loss were animals that did not recover from anesthetic during

44

surgery or one of the weekly radiographs. The final table of specimen number is

given in Table 2.4-1.

Table 2.4-1: Specimen sample size per group

6 weeks 9 weeks 12 weeks 18 weeks Allografts 6/11 6/13 13/14 11/11 Autografts 7/11 12/14 13/14 7/11

2.4.2 µCT-based morphology and structural indices

Representative micro-CT sagittal-sections of typical autografts and allografts

demonstrate several remarkable differences in the size and morphology of the new

mineralized callus and graft bone over time (Figure 2.4-1). The prominent differences

between the grafts at 6 weeks are: (1) the large callus that envelops the entire length

of the autograft but remains limited to the ends of the allograft, (2) the clear union of

the autograft to the proximal and distal junctions of the host femur versus the limited

connectivity observed in the allograft, and (3) the differences in graft remodeling over

time. By 9 weeks the creeping callus covered large segments of the allograft,

resulting in a marked increase in union between the unresorbed allograft and the host.

This extensive remodeling continued over time, such that at 12 and 18 weeks the

autograft bone was unidentifiable in many of the samples, and the new bone collar

was the only contiguous load-bearing bone in the mid-femur. In contrast, allografts

did not display evidence of resorption up to 12 weeks postgrafting. However, the

45

callus at the allograft–host interface remodeled progressively over time and appeared

to establish limited graft–host continuity by 18 weeks.

Figure 2.4-1: Micro-CT images of allografts and autografts healing over time.

Mice receiving live autografts or devitalized allografts to repair segmental femoral

defects were sacrificed at 6, 9, 12, and 18 weeks following surgery and their grafted

femurs were analyzed by micro-CT. A representative image from each time point is

shown. Of note are the dramatic differences in osteointegration at 6 weeks (white

46

arrows), new bone collar formation around the graft at 6 and 9 weeks (red arrow

heads), and graft (shaded in yellow) remodeling such that only allografts are much

more intact at 12 and 18 weeks. [Reproduced from Reynolds 2007 with permission

of the American Society of Bone and Mineral Research]

Although no statistically significant differences were observed in BVCallus

between the grafts (Figure 2.4-2), the data demonstrated a trend in which the

autografts were on average 1.15 to 1.66-fold larger than the allografts. By contrast,

BVGraft of the autografts were only 62% of the allografts (p<0.01) at 6 weeks, and

declined progressively over time reaching 44% and 40% of the allograft BVGraft at 9

and 12 weeks, respectively. As demonstrated in Figs. 2.6 and 2.7, allograft BVGraft did

not change significantly over time up to 12 weeks. However, remodeling over time

made it difficult to clearly define both graft types at 18 weeks in many of the samples,

such that their BVGraft were similar at 18 weeks (Figure 2.4-2).

47

6 9 12 180.0

0.2

0.4

0.6

0.8

AllograftAutograft

Time (Weeks)

BV C

allu

s[m

m3 /m

m]

6 9 12 180.0

0.2

0.4

0.6

0.8

1.0

1.2

***

******

Time (Weeks)

BV G

raft

[mm

3 /mm

]

Figure 2.4-2: Mineralized callus and graft volumes during femoral allograft and

autograft healing over time.

The calcified callus volume BVCallus (A) and the bone graft volume BVGraft (B), at each

time point, normalized to each sample’s graft length. Data represent means ± SEM.

Asterisks indicates significant differences between autografts and allografts at each

time point (*** p<0.001) [Reproduced from Reynolds 2007 with permission of the

American Society of Bone and Mineral Research]

Furthermore, the only significant difference in PMI between the two grafts

was at 6 weeks when the autografts’ PMIMid Graft was 2.1-fold greater than allografts’

48

(p<0.001; Figure 2.4-4). This reflects the massive amounts of bridging callus

surrounding autografts, versus the naked cortex of the allograft at 6 weeks.

Allograft Autograft

Figure 2.4-3: Cross-sectional PMI Analysis of a representative allograft and

autograft.

Representative surface renderings of an allograft (top left) and an autograft (top

right) at 6 weeks showing the location and morphology of selected coronal sections

through the grafted femurs. Of note are the remarkable differences in cortical bone

thickness and the area of the femoral slice between the two graft types. Those slices

are indicated on the PMI line graphs (bottom) by gray triangles. The dotted vertical

lines define the proximal and distal ends of the graft. The minimum PMI (PMIMin)

over the graft region is indicated by *, and PMIMax is indicated by #. The average

PMI over the middle 50% of the graft (PMIMidGraft and the average PMI at the

proximal and distal ends of the grafted region (PMIGraftEnds) were also determined for

each specimen. [Reproduced from Reynolds 2007 with permission of the American

Society of Bone and Mineral Research]

49

0.0

0.5

1.0

1.5

6 wks 9 wks 12 wks 18 wks

PMI-A

ve. (

mm

4 )

*

0.0

0.5

1.0

1.5

2.0

2.5

3.0

6 wks 9 wks 12 wks 18 wks

PMI-M

ax (m

m4 )

*

0.0

0.5

1.0

1.5

6 wks 9 wks 12 wks 18 wks

PMI-M

in (m

m4 ) *

**

0.0

0.5

1.0

1.5

2.0

2.5

6 wks 9 wks 12 wks 18 wks

PMI-G

raft

Ends

(m

m4 )

*

0.0

0.5

1.0

1.5

6 wks 9 wks 12 wks 18 wksPM

I-Mid

Gra

ft (m

m4 )

*

**

A CB

D E

AllograftAutograft

Figure 2.4-4: Cross sectional Polar Moment of Inertia Data.

The cumulative PMIAve (A), PMIMax (B), PMIMin (C), PMIGraft Ends (D) PMIMidGraft (E) are

presented as mean ± SEM . * indiciate statisitically significant differences between

allograft and autografts. [Adapted from Reynolds 2007 with permission of the

American Society of Bone and Mineral Research]

2.4.3 Biomechanical testing

At 6 weeks, the ultimate torque and torsional rigidity of autografts were 2.5

and 3.9 times greater than allografts (p<0.001 and p<0.0001, respectively, Figure

2.4-5). Remarkably, by 9 weeks the differences between autografts and allografts

were insignificant, and the ultimate torque and torsional rigidity of both graft types

were not different than age-matched unoperated femurs (20.23 ± 2.28Nmm and

922.18 ± 103.40 Nmm2/rad, respectively). However, between 9 and 18 weeks,

50

allografts experienced a significant 45% reduction in ultimate torque (p<0.0005). The

torsional rigidity of the allografts also decreased significantly by 45% between 9 and

12 weeks (p<0.0005), and then increased significantly by 41% (p<0.05) at 18 weeks.

By contrast, the torsional properties of autografts did not change significantly

(p<0.05) between 9 and 18 weeks.

6 9 12 180.0

5.0

10.0

15.0

20.0

25.0 AllograftAutograft

*** ****

A

Time (W eeks)

T Ult

[ N.m

m ]

6 9 12 180

250

500

750

1000

**

***

B

Time (W eeks)

TR[ N

.mm

2 ]

Figure 2.4-5: The torsional properties of femoral allografts and autografts over

time.

Following micro-CT imaging, grafts were tested in torsion at a rate of 1o/s until failure

and the (A) ultimate torque (TUlt), and (B) torsional rigidity (TR), were computed as

described in the methods. Asterisks indicate significant differences between

51

autografts and allografts for that time point (*p<0.05, **p<0.01, and ***p<0.001). Data

represent mean ± SEM. [Reproduced from Reynolds 2007 with permission of the

American Society of Bone and Mineral Research]

Analysis of the three modes of failure described in Figure 2.3-4 are given in Table

2.4-2 as the percentage of incidence of the different modes. The critical χ2 value for α

= 0.05 with 6 degrees of freedom is 12.59. Since the χ2 for this table is 22.92 which

is greater than χ2critical, the pattern of allograft failure is statistically different than

autografts (p<0.005), as nearly 64% of allografts fail in Pre-union or Early union

modes even after 12 weeks of healing, and more than 70% of autografts failed in

Mature union mode 12 and 18 weeks post-grafting.

Table 2.4-2: Distribution of allograft and autograft modes of failure in torsion over

time

Allografts† Autografts

Week Pre-union Early Union Mature Union Pre-union Early Union Mature Union

6 100% – – 66% 33% –

9 75% 25% – 36% 28% 36%

12 28% 36% 36% – 29% 71%

18 18% – 82% 29% 71%

2.4.4 Correlations between micro-CT parameters and torsional properties

To the end of identifying micro-CT-derived parameters that correlate with

biomechanical strength of structural bone graft in our mouse model, we first

52

performed univariate linear regression analyses. Unfortunately, although significant,

these regression analyses failed to identify any strong correlations (Table 2.4-3). To

improve the predictive power of these correlations we performed multivariable

regression analyses using the best subset selection method. These tests demonstrated

significant correlations between combinations of bone volume and cross-sectional

PMI with biomechanical properties (Figure 2.4-6). The ultimate torque was best

predicted by a combination of BVTotal, BVCallus, PMIMin, and their interactions with

time (Adjusted R2 = 0.44, Figure 2.4-6 A, B). The torsional rigidity was best

predicted by a combination of the micro-CT variables PMIAverage, PMIMin, PMIGraft Ends

and their interactions with time (Adjusted R2 = 0.50, Figure2.4-5 C, D).

Table 2.4-3:Correlation coefficients and significance levels for single variable linear

regression models of micro-CT-based estimates of graft ultimate torque and torsional

rigidity

Ultimate Torque Torsional Rigidity Measure †Relationship †R2 §P Relationship R2 P BVCallus 0.09 <0.01 0.005 N.S. BVGraft (–) 0.08 <0.05 (–) 0.18 <0.001PMIAverage 0.06 <0.05 (–) 0.05 <0.05 PMIMax 0.001 N.S. (–) 0.13 <0.005PMIMin 0.32 <0.0001 0.024 N.S. PMIMidGraft 0.21 <0.0001 0.000 N.S. PMIGraft Ends 0.001 N.S. (–) 0.14 <0.001

† Negative symbols indicate inverse linear correlations.

53

§ P values indicate the significance level of the hypothesis that the slope of the

regression line is different than zero. N.S. indicates P > 0.05

Intercept 15.55 ± 4.53* Time -0 ± 0.33 BVCallus 11.59 ± 8.25BVTotal -7.35 ± 1.78* PMIMin -0.47 ± 5.90 Time × BVCallus -0.69 ± 0.69 Time × PMIMin 1.41 ± 0.53*

.28

Intercept 187.06 ± 240.32 Time 14.18 ± 16.39PMIAverage 1009.35 ± 434.10*PMIMin -480.08 ± 363.89PMIGraft Ends -753.66 ± 336.99*Time × PMIAverage -161.34 ± 47.42*Time × PMIMin 153.44 ± 39.59*Time × PMIGraft Ends 92.79 ± 37.83*

A B

CD

Figure 2.4-6: Multivariable linear regression analysis micro-CT parameters vs

mechanical outcomes.

Multivariable linear regression analysis based on the best subset selection of

geometric micro-CT-based parameters including bone volume and PMI, and their

interactions with time show strong correlations between the predicted and

experimentally measured ultimate torque (A) and torsional rigidity (C) independent of

the graft type. The dotted diagonal lines represent the ideal case of perfect

54

prediction. Tables (B) and (D) list the independent variables and their coefficient

estimates which were selected. Data represent parameter estimate ± standard error

* Indicates p<0.05 [Adapted from Reynolds 2007 with permission of the American

Society of Bone and Mineral Research]

0 200 400 600 800 1000

Torsional Rigidity  (N*mm2)

Allografts

Autografts

0

1

2

3

0 10 20 30

Ultimate Torque (N*mm)

Allografts

Autografts

Pre-union

Early union

Mature union

Figure 2.4-7: Torsional strength and rigidity vs mode of failure.

The mode of failure, as determined by post-torsion testing x-rays versus the ultimate

torque and torsional rigidity for allografts and autografts at 6 and 9 weeks.

2.5 DISCUSSION

While it has long been recognized that the biomechanical properties of

structural autografts are superior to allografts, the mechanisms responsible for these

differences and when they are manifested during the healing process are poorly

understood. Moreover, there are no noninvasive imaging-based outcome measures to

assess the biomechanical properties of structural grafts, such that potential adjuvants

that improve allograft healing could be evaluated without destructive biomechanical

55

testing. Before such outcome measures can be translated to clinical applications, they

would first have to be developed and validated in pre-clinical animal models.

To address these issues, we aimed to characterize the 3D morphological and

biomechanical differences between allograft and autograft healing over time, and to

identify the micro-CT parameters that best correlate with torsional strength and

rigidity using a mouse femoral graft model. Consistent with previous studies

(Tiyapatanaputi et al. 2004), we found that autografts display a robust osteogenic

response that was previously reported to derive from the periosteum of the donor and

host bone (Zhang et al. 2005; Zhang et al. 2005), which results in a new bone collar

that completely surrounds the graft by 6 weeks. In contrast, allografts heal via

creeping callus from the host that remains scant until 9 weeks of healing. This may

have contributed to the dramatic difference in ultimate torque to failure and torsional

rigidity between the devitalized femoral allografts and autografts at 6 weeks (Figure

2.4-5).

Analysis of the micro-CT images delineated three observations which may

explain how allografts achieve biomechanical equivalency to autografts at 9 weeks

via different mechanisms of healing. The first observation is the very large callus at

the allograft ends at 6 weeks which remodels down over the next 3 weeks and slowly

creeps onto the allograft cortical surface, resulting in an increased PMIMid Graft at 9

weeks (Figure 2.4-1 and Figure 2.4-4). In contrast, autografts are enveloped by a

bridging callus with a thin cortex as early as 6 weeks which results in a large and

somewhat uniform PMI throughout the length of the graft and subsequently remodels

56

down uniformly over time. The second is the dramatic difference in cortical bone

thickness between the two graft types which results from uneven rates of graft

remodeling as the autografts experience rapid and robust resorption (Figure 2.4-1).

The third is the union of the graft with the host bones which was qualitatively

deduced a priori from the observed modes of failure following the torsion testing.

These data indicate that all grafts were poorly connected to the host at 6 weeks as

100% of the 6- week allografts failed in Pre-union compared to only 66% of the 6-

week autografts (Table 2.4-2). Interestingly, about 20% of the allografts never

achieved the Early union stage even after 18 weeks of healing, suggesting a persistent

poor incorporation of the allograft with the host. It should be noted that the failure

classification of Pre-union should not be interpreted to indicate that there was

absolutely no union in the specimens failing in this mode. Indeed all the tested

specimens registered torsional resistance that varied with graft type and time post-

grafting as shown in Figure 2.4-5. Naturally, a quantitative measure of the degree of

union between the graft, the callus, and the host would likely be an important

determinant of the biomechanical stability of the graft. To our knowledge there are no

existing methods that have been published for quantitatively assessing the level of

connection between complicated bone segments in 3D. Studies that have performed

other analyses similar will be reviewed in Chapter 3, along with an explanation of the

limitations of those methods. We therefore began working on developing methods to

determine a quantitative 3D parameter from micro-CT image data. The methodology

that was settled upon is explained in Chapter 3.

57

The causes for autograft resorption may start from the transfer of load-sharing

from the graft to the newly formed external callus which bridges from host-to-host,

thus shielding the autograft from stress which could cause bone disuse atrophy.

Overall, the autografts had greater ultimate torque and torsional rigidity

compared to the allografts over time (Figure 2.4-5). However, this was not

necessarily due to a delay in allograft healing, as the biomechanical properties of

autografts and allografts were equivalent by 9 weeks. At 12 and 18 weeks, the

significant decreases in allograft torsional rigidity and ultimate torque seem consistent

with clinical observations that patients receiving massive structural allografts are

initially able to return to normal activity and function but experience allograft failure

1–2 years after implantation (Lord et al. 1988; Berrey et al. 1990; Wheeler et al.

2005). While the cause of this late stage failure is not fully understood, the

accumulation of microdamage and the initiation of resorption in devitalized allografts

are likely factors in graft weakening that need further investigation.

While two-dimensional radiographic measurements (X-rays) have been the

standard clinical technique to assess the progress of bone fracture healing, they have

been shown to be poor predictors of fracture strength and rigidity (Nicholls et al. 1979).

Other imaging modalities including dual-energy X-ray absorptiometry (DXA) have

shown that the callus bone mineral content and density in experimentally induced

cortical bone fracture or osteotomy models correlate significantly with torsional

strength and rigidity of the fractured bones (Markel et al. 1991; den Boer et al. 1999;

Blokhuis et al. 2000) and implanted autografts (Delloye et al. 1986). However, in

58

more complex situations such as devitalized cortical allografts that are known to

exhibit delayed and impaired remodeling and turnover, bone mineral content and

density measurements can be misleading and are likely to be poor predictors of graft

strength and incorporation.

Unlike conventional X-ray and DXA that offer low resolution information

about the 2D geometry and mineralization of bone fracture or graft callus, computed

tomography imaging provides 3D details about the callus geometry and volume. In

our study, univariate correlations between micro-CT parameters and the torsional

properties yielded weak or insignificant correlations that at best explained no more

than 32% of the variability in the biomechanical properties of the mouse femoral

grafts (Table 2.4-3). These results are consistent with reports demonstrating that

estimates of healing progress based on bone volume and PMI measurements from CT

images of fractured rat femurs had weak and insignificant associations that explained

no better than 9% of the measured fractured femoral torsional rigidity (Shefelbine et

al. 2005). The multivariate correlations that were generated in the mouse femoral

graft model were as good as those obtained using sophisticated CT-based finite

element (FE) prediction of torsional and flexural femoral strength in a rat femur

fracture model (Shefelbine et al. 2005). While the use of FE modeling to clinically

predict fracture risk has only seen limited applicability in the reported literature and

provided only anecdotal data on only one patient (Taddei et al. 2003), direct statistical

interpretation of data from CT imaging may have wider clinical applicability in the

assessment of grafted bone healing (Manfrini et al. 2004).

59

One of the potential limitations in translating this work to clinical applications

is that clinical CT imaging systems sacrifice the quality and resolution for faster scan

acquisition times and reduced radiation exposure. Therefore, the low clinical CT

scanner resolution may limit the direct extrapolation of our micro-CT correlates to the

clinical setting. The state of the art clinical CT scanners can routinely achieve

comparable relative in-plane resolution but out-of-plane resolution is less than

optimal. Table 2.5-1 compares the relative resolution of the micro-CT system against

the current standard of care scanner. In-plane resolution of clinical imaging is

equivalent to micro-CT imaging of mouse cortical bone, but is much worse out-of-

plane because slice thickness is 2-8 times larger than the in-plane pixel dimensions.

Further, standard clinical CT imaging is unable to resolve individual human

trabeculae as well as micro-CT can in mice. A more thorough analysis of the effects

of the lower resolution on the graft and callus volume computation and the predictive

correlations with biomechanical properties would have to be performed in large

animal preclinical and future human clinical studies. Regardless, there are clinical

reports that strongly suggest the value of CT imaging, despite the low resolution

limitations, in the management of massive long bone allografts(Manfrini et al. 2004;

Attias et al. 2006). However, these limited studies do not report any quantifiable

correlations between CT parameters and functional recovery, which underscores the

importance of delineating such correlations in the preclinical mouse femoral graft

model as a first step towards clinical translation. Additional discussion of the need for

preclinical and clinical translational studies is included in Chapter 5.

60

61

Table 2.5-1: Relative resolution of routine clinical CT images to micro-CT images for

cortical bone and trabecular bone.

Mouse Human Relative resolution Human clincal scan

vs. Mouse µCT

Femoral cortical

Femoral metaphyseal trabecular

Humeral cortical

Proximal tibia trabecular cortical trabecular

Thickness (µm) 180a 41[32b - 49c] 5000d,e 123 [76g -170f]

Res

olut

ion

(µum

) In-plane 12.2 [10.5 – 13.9] 310 [230 – 390]

Out of plane 12.2 [10.5 – 13.9] 1250 [625 – 2500]

Pixe

ls p

er

thic

knes

s In-plane 14.8 3.4 16.1 0.4 1.02 0.12

Out of Plane 14.8 3.4 4 0.1 0.27 0.03

Sources: a (Brodt et al. 1999), b (Martin-Badosa et al. 2003), c (MacDonald et al.

2007), d (Tingart et al. 2003), e (Thiele et al. 2007), f (Day et al. 2000), g (Boutroy et

al. 2005). The median value for dimension and resolution was used, and the range

is given in brackets.

62

Chapter 3:  Development of an algorithm for the quantification of graft­to­host union using micro­CT imaging  3.1 INTRODUCTION

The ability to evaluate patient outcomes longitudinally after bone grafting is

limited by a paucity of available non-invasive outcome measures. This limits the

ability to identify and treat patients with complications before they suffer a traumatic

failure. This also limits our ability to study adjuvant treatments quantitatively and to

determine differences in efficacy which would be necessary for ranking treatment

options based on performance, and for optimizing the best treatments such as dosage

and delivery method. In the study described in Chapter 2, two treatment groups

(allografts and autografts) were compared as a basis for identifying which of the

available outcome measures were most successful at predicting the best case scenario

for healing. These outcome measures included quantitative graft and callus bone

volume measurements, and the maximum, minimum and average cross sectional

polar moments of inertia. Together in a multivariate regression equation, these

parameters were only able to is result was only able to predict up to 44 and 50% of

the variation in the torsional strength and rigidity, respectively, which suggests that

these CT variables fail to explain about half of the variability in the torsional

biomechanics. Based on the observation in Figure 2.4-1 that non-unions between the

graft and host were present, but unaccounted for in the correlations of Chapter 2,

along with the result that the mode of failure after biomechanical testing often

63

occurred at the graft-host interface (Figure 2.3-4, Table 2.4-2) suggesting that was the

weakest link, we therefore hypothesized that the graft-to-host union would correlate

significantly with torsional biomechanical properties. The absence of a non-invasive,

objective, 3D measure of the degree of union makes studying this problem

impossible.

We chose to study allografts and autografts at 6 and 9 weeks because we

hypothesized that these early time points had the greatest variation in the degree of

union based on observations of CT images and post-testing analysis of the mode of

failure indicated that non-unions were prevalent among allografts at 6 weeks, but by 9

weeks they had established union to a greater degree and were almost 3x stronger and

5x more rigid. At later time points, mouse bone healing was substantial which

resulted in few apparent non-unions from radiography, and due to some extensive

remodeling, graft bone was somewhat indistinguishable from the callus making it

difficult to determine what was graft and what was host, and therefore how they were

actually connected. Therefore we limited our analysis to specimens for which the

degree of union was able to be obtained, as well as specifically investigating a time at

which our model seemed to be recapitulating a known deficiency in clinical graft

healing.

3.1.1 Previous attempts at quantifying union

Despite the understanding that non-unions cause instability and in the case of

reconstructive grafting, a delay in union means prolonged loading of fixation

instrumentation, there is no way of quantifying the degree of union. Two dimensional

64

radiographic evaluation has been performed to qualitatively assess whether cortices

had bridged on anterior-posterior and/or medial-lateral x-rays (Brown et al. 2004).

Brown et al. were unable to achieve any statistical significance among three treatment

groups harvested at 3 time points in a standardized rat femoral fracture model. This

was adapted to 2D planar CT slices by Gardner in 2007 to again qualitatively assess

the bridging of cortices in 4 sampled sites in a mouse tibia study, but again was

unsuccessful at delineating treatment groups. This negative result could be due to

limited sampling of only two slices per specimen, as the range with the number of

bridged cortices varied from 0 to 3 for all groups in the study. Bressler attempted

quantitative automated analysis of x-rays of fracture union sites by quantifying the

mineral density at the graft-host interface of distal femoral allografts in sheep using

digital image analysis of x-ray films. They found that this improved classification of

unions and non-unions over trained observers (Bresler et al. 1998).

To determine the osteoconductive effects of porous poly(propylene fumarate)

(PPF) foam coating on the integration of intercalary cortical (tibial) allografts in a rat

model, Lewandrowski et al. described a histomorphometry technique for assessing

allograft-host integration by tracing the perimeter of the graft and estimating the

length of host-to-graft integration along that perimeter (Lewandrowski et al. 2002).

The percentage of length that was connected was determined independently for both

the proximal and distal ends of each graft and named the Bonding Index.

Unfortunately, this method is limited by the destructive nature of this histological

evaluation that does not permit one-to-one correlations of graft-host union with

65

biomechanical properties. We thought we could advance this 2D technique by

extending it to 3D to eliminate sampling error, using non-destructive imaging to

measure union without compromising the specimen.

In this chapter, a novel micro-CT based algorithm is described and validated

which computes a 3D measure of union between host (bone and callus) and mouse

femoral graft (autograft or allograft) based on the surface area of the graft onto which

bone forms to connect the graft to the host. The ratio of connected graft area to total

graft surface area is computed for each graft end and the lesser value for each graft is

termed the Union Ratio. This technique is then used to investigate the variation in the

osseointegration of mouse grafts over time to test the hypothesis that the Union Ratio

correlates significantly with the torsional strength and rigidity of bone allografts.

Lastly, the translational potential of this measurement technique is briefly

demonstrated using clinical CT data of a nonunion tibia fracture to show that despite

lower relative resolution (as indicated in Table 2.5-1), this technique could be useful

and stimulates further validation in future studies.

3.2 METHODS

3.2.1 Experimental model

Specimens analyzed in this study are a subset of the 4mm intercalary femoral

allograft and autograft study described in Chapter 2. This included allografts (n=7)

and autografts (n=8) at 6 weeks and allografts (n=12) and autografts (n=7) at 9 weeks

66

after surgery. The same outcome measures from the first study were used, including

bone volumes, polar moments of inertia, torsional rigidity and strength.

3.2.2 Union Ratio Algorithm

A custom software program named uCTAnalysis was written in MATLAB

(The Mathworks, Natick, MA) for the analysis of the Union Ratio from the micro-CT

images. An active contouring algorithm (Kass et al. 1988) was adapted for the semi-

automated generation of a shell around the graft. First, contours are drawn around the

periosteal and endosteal surfaces of the bone graft in a single transverse micro-CT

slice which has been lightly low-pass filtered using a 2D Gaussian filter (σ = 1.8

pixels; Figure 3.2-1A, B). The contour then snaps to the edge of the graft based on

the 2D gradient of the grayscale image using a Prewitt filter (Prewitt 1970) (Figure

3.2-1C). Lastly, the contour dilates to a neighboring pixel with the darkest grayscale

intensity along a 4 pixel-long line drawn from each contour point perpendicular to the

line that connects the two contour points on each side of the current contour point.

Thus the contour dilates into the void space between the graft and callus bone if it

exists (Figure 3.2-1D). Because the contour snaps to the gradient between

contrasting pixels, the contour point will shift to the material of lesser radiopacity.

Generally, this means it shifts off the dense cortical graft, onto either newly

mineralized callus (woven bone is less radiopaque than organized lamellar bone), or

onto unmineralized soft tissue adjacent to the graft. Cubic spline interpolation was

used to smoothly join the contours. The contour from the previous slice is then copied

67

onto the next where the edge-detection and void space search processes are repeated

under operator supervision and modification, until the entire length of the graft is

contoured to create a shell around the graft. The shell is then meshed using triangular

elements that are used to quantify the amount of graft area in contact with host bone

or mineralized callus by summing 1/3 of the area of each triangle element for each

vertex that falls within a voxel with a grayscale value greater than the threshold used

to define mineralized tissue. The proximal and distal halves of the graft are evaluated

separately, and the lesser ratio of the connected surface area to total graft surface area

is used in the analysis and assigned as the value of the Union Ratio, to account for

any variation in graft size in our standardized model. The MATLAB code for the

semi-automated contouring, the 3D shell generation and the quantification are given

in the appendix and on a CD.

A B

C D

E

Figure 3.2-1: Illustration of the graft-to-host Union Ratio algorithm.

68

A user outlines the surface of the graft using contours on transverse micro-CT slices

(A). The semi-automated algorithm developed using MATLAB then optimizes the

manually defined contours drawn around the endosteal and periosteal surfaces

(yellow lines) (B). The contours are first snapped to the graft boundary by edge-

detection (C), then dilated into darker regions away from the graft surface, finding the

gap between graft and callus, if it exists (red denotes voxels that are adjacent to host

bone/callus and blue denotes voxels that are adjacent to host soft tissue) (D). The

resulting 2D contour from one slice is then copied to the next slice and the edge

detection and gap-finding operations are performed. This process is repeated on

each slice until the entire graft is enclosed in contours. A smoothed 3D shell is

generated from the contours using MATLAB’s isosurface function (E). The footprint

of bone penetrating the shell therefore defines connection areas between the graft

and host or callus. Summed over the entire surface area of either half of the graft

shell, the lesser area of the connections (red regions) normalized by the total surface

area for either proximal or distal half is defined as the Union Ratio.

3.2.3 Validation with Digital phantom

A digital model with standard hollow cylindrical geometry was created in

order to validate the calculations used to measure the connected surface area. This

model was generated as an idealized graft between two host ends, with geometrically-

defined connections simulating callus originating from the host tissue. The

theoretically-predetermined areas of connectivity that were used to validate the

computational technique are shown in Figure 3.2-2. The hollow cylindrical model

69

was generated with thickness of 15 pixels and outer diameter of 50 pixels, yielding a

relative resolution similar to the resolution of the real micro-CT images (typical

allograft cortical thickness was 180-200 microns (13-14 pixels) thick, and about 1.25-

1.55 mm (90-110 pixels) in diameter. Predetermined areas of connectivity were

created directly between the graft and host as rectangular prisms that either attached

to the end surface of the graft (Figure 3.2-2 B-B) or intersected the periosteal surface

of the graft connecting it to the callus (Figure 3.2-2 C-C). This idealized model was

then contoured and the Union Area was computed as described for the experimental

grafts.

B

B

C

C

Section B-B Section C-C

A

D

Figure 3.2-2: Algorithm validation using a digital model.

An idealized cylindrical graft (blue) between host cortical bone (white) and callus

(light gray) was digitally generated in MATLAB and used to validate the Union Ratio

70

measurement. The graft was given defined rectangular regions of union to the host

directly (section B-B) as well as between the graft and the callus forming around it

(section C-C). The theoretical union area (red regions) based on the idealized

geometry projected onto the curved surface was 2173.2 pixels2. Using the

contouring computational algorithm, the measured area represented in (D) was

2171.4 pixels2 resulting in a measurement error of only 0.08%.

3.2.4 Statistical Analysis

Comparisons of autograft and allograft Union Ratio data at the different time

points were performed using 2-way analysis of variance and Bonferroni post hoc

multiple comparisons. To evaluate intraoperator and interoperator error in the

estimation of the Union Ratio, a subset of 2 specimens from each group (8 specimens

total) was randomly selected to be repeated by the first operator (DGR) as well as

performed and repeated by another trained operator (MOP). The average percent

error between measurements was calculated by the absolute value of the difference

between measures divided by the average measurement. As described by Lodder et al

(Lodder et al. 2004) the coefficient of variation (CV) is the standard deviation

between measurements normalized by the mean of the paired measurements,

calculated as where a and b are the first and the second

measurements, Ma and Mb are the mean values for the two groups, and n is the

number of paired observations. Intraclass correlation coefficients (ICC) to evaluate

the concordance, or agreement, between measurements within and between operators

71

were computed (McGraw et al. 1996). This is defined as the difference between the

overall variation and the measurement variation, divided by the sum of the

measurement and overall variation. The ICC ranges between 0 and 1 where 1 is

perfect concordance.

As described in Section 2.3.5, univariate regression analysis was used to

determine if Union Ratio correlated with the ultimate torque and torsional rigidity of

6 and 9 week specimens. The best single variable regressions were determined for

the allografts and autografts separately as well as together using MiniTab Release 12

(State College, PA). Multivariate regression analysis was used to determine

combinations of micro-CT parameters that correlated with the torsional mechanical

properties using SAS 9.1 (SAS Institute Inc., Cary, NC). The analyses were

performed 3 separate ways. The foundation set included the variables that were used

in Chapter 2 (BVs and PMIs). In the second evaluation the Union Ratio was included

as a covariate. Interactions between the Union Ratio and the other foundation

variables were also allowed in this regression analysis. The third evaluation included

time as a covariate and investigated its interactions with the foundation variables. In

each evaluation, in order for two interacting variables to be included as a term, they

also needed to be included in the regression by themselves. This is accomplished by

creating variable groups in SAS so that interacting terms are only selected if the two

variables that make up the interacting term are also included, and together the three

terms significantly add to the regression. Unfortunately, SAS cannot not perform best

subsets selection using Mallow’s CP with grouped variables; instead, bi-directional

72

stepwise selection was used to select significant terms for the model. The selection

criteria used for this was to add the next strongest variable in the regression if it

achieved a significance at p < 0.50 to enter the model, and then check the significance

of each term of the equation and remove the one with the lowest significance if p <

0.05. These steps are iterated to ensure the most significant independent variables

and variable-interactions would be included in the model. The limitation of switching

to stepwise regression is that it does not look at every combination of independent

variables as Best Subsets selection does, so it could miss a better combination of

parameters, but in our experience evaluating both methods this risk is generally small

and the methods give similar results.

A continuous host-to-host callus shell had formed from one end of the graft to

the other in some specimens, while in other specimens there was a noticeable

discontinuity between the callus that formed from the proximal and distal ends. Host-

to-host bridging was particularly apparent in many of the autograft specimens.

Characterization of specimens with or without continuous host-to-host callus

formation was systematically investigated by scanning the axial cross sections from

one end of the graft to the other. If there was an interruption in callus, they were

“not-bridged”, while those with continuous flow of callus from one end to the other

were termed “bridged”. Non-bridged specimens are apparent in Figure 2.3-2C and

the allografts at 6 and 9 weeks in Figure 2.4-1 with all other specimens shown having

bridging (the 18 week allograft specimen shown had bridging apparent in another

73

view). To examine this as a factor for predicting strength, it was also incorporated in

the regression analysis.

3.3 RESULTS

3.3.1 Algorithm Validation

To validate the semi-automated contouring algorithm and computation of the

Union Ratio, a digital model was created that resembles a graft connected to host

bone and callus by a footprint of defined dimensions (Figure 3.2-2). The

predetermined connected area which accounts for the curvature of the cylindrical

model surface was computed to be 2173.2 pixels2. Using the contouring method and

the MATLAB algorithm, the Union Area was determined to be 2171.4 pixels2,

resulting in a measurement error of only 0.08%.

3.3.2 Union Ratio of Autografts and Allografts

Figure 3.3-1 illustrates the typical differences in the union with host bone and

callus between allografts and autografts at 6 and 9 weeks. Table 3.3-1 reports the

cumulative group results. At 6 weeks, the Union Ratio of autografts was nearly

double that of allografts (p < 0.05). The areas of union were also more uniformly

distributed along the length of the autografts compared to the allografts for which

new bone formation was restricted to the host bone at the ends of the grafts (Figure

3.3-1). At 9 weeks, the allografts’ Union Ratio was 2.2 times that of 6 week

74

allografts (p < 0.05), while the autografts’ Union Ratio declined 33% from 6 to 9

weeks (p < 0.05).

6wk Allo 9wk Allo 6wk Auto 9wk Auto

0.05 0.27 0.28 0.13

Figure 3.3-1: Representative micro-CT and union area images.

Representative micro-CT sagittal sections of 6 and 9 week allografts and autografts

(top) with the corresponding union area maps and Union Ratio numerical values

(bottom). The graft bone is highlighted in yellow. Red indicates areas where the graft

is connected to the host. Note that the areas of union and non-union (green arrows)

correspond accurately to the measured union areas on the surface of the graft

represented in red. Note also that union with the periosteal and endosteal surfaces,

and the ends of the graft were all accounted for. The proximal and distal halves of

75

the graft were evaluated separately, and the lowest value of the union area

normalized by the surface area was reported as the Union Ratio underneath each

specimen. The Union Ratio for each specimen is given at the bottom (numerical

values).

Table 3.3-1: UnionRatio and Host-to-Host bridging callus results

6 wk Allo 9 wk Allo 6 wk Auto 9 wk Auto

Group: n=8 n=7 n=8 n=12

UnionRatio: 0.105 (0.023) 0.228 (0.037) * 0.224 (0.029) † 0.150 (0.011) *,†

H-H Bridg.: 0% 57% 100% 100%

TUlt 5.0 (1.2) 14.4 (0.9) * 17.1 (1.2) † 18.4 (1.1) *,†

TR 0.105 (0.023) 0.228 (0.037) * 0.224 (0.029) † 0.150 (0.011) *,†

Data presented as mean (SEM) and as percent of specimens with bridging. Significantly

different means are labeled † for p < 0.05 between time points for each graft type and * for p

< 0.05 between graft types at each time point. TUlt and TR are the same results as presented

in Chapter 2.

The intra- and inter-operator sources of error in the measurement of the Union

Ratio were also investigated. The average percent error between operators’

measurements was 12% and the coefficient of variation (CV) was 9.7%. The intra-

operator ICC was 0.930 for DGR and 0.949 for MOP while ICC between different

operators (DGR and MOP) was 0.926. These results indicate that the measurements

were remarkably reproducible.

76

3.3.3 Correlations between Union Ratio and Torsional Properties

To estimate the effects of the Union Ratio on the torsional biomechanical

properties, univariate linear regression analyses were performed. When autografts

and allografts at all time points were grouped, the regression analysis identified weak,

yet significant, associations between the Union Ratio and the torsional properties

(Table 3.3-2). However, when analyzing the allograft data separately the correlation

was much stronger. By contrast, there were no significant associations between the

autografts’ Union Ratio and torsional properties. Taken together, these results suggest

that the Union Ratio is a significant indicator of functional strength in the devitalized

allografts that undergo no or little remodeling over the first 9 weeks of healing, while

it does not correlate with the biomechanical properties of autografts that undergo a

robust remodeling (as shown in Figure 2.4-2) such that the Union Ratio actually

decreases between 6 and 9 weeks due to excessive graft resorption.

77

Table 3.3-2: Coefficients of determination and p-values for the univariate linear

regression of non-structural and structural independent variables TUlt and TR.

Allografts & Autografts Allografts Autografts

TUlt TR TUlt TR TUlt TR

Non-structural

Time 48.1 (0.0001)

65.1 (0.0001)

80.2 (0.0001)

97.4 (0.0001)

27.3 (-) (0.022)

33.7 (-) (0.005)

Graft Type 25.0 (0.002)

9.0 (0.09) n/a n/a

Structural

UR 12.1 (0.043)

15.4 (0.022)

58.4 (0.001)

50.7 (0.003)

15.1 (0.1)

4.6 (0.379)

BVCallus 1.9

(0.44) 0.2

(0.812) 8.1

(0.304) 4.7

(0.44) 2.0

(0.567) 0.4

(0.803)

BVGraft 36.4 (-) (0.0001)

16.7 (-) (0.016)

12.6 (0.194)

5.8 (0.388)

19.1 (-) (0.061)

27.0 (-) (0.023)

PMIAve 9.5

(0.075) 11.9 (-) (0.045)

20.0 (0.094)

15.1 (0.152)

4.7 (0.375)

2.0 (0.561)

PMIMax 23.9 (-) (0.003)

22.6 (-) (0.004)

35.0 (-) (0.020)

27.3 (-) (0.046)

18.0 (0.070)

0.1 (0.916)

PMIMin 16.8

(0.027) 7.9

(0.108) 7.1

(0.336) 16.4

(0.135) 0.9

(0.701) 5.5

(0.334)

H-H Bridging 57.2 (0.0001)

35.5 (0.0001)

61.3 (0.001)

49.1 (0.004) n/a *

Data presented as R2 (p < ). P values for the two-sided test of the null hypothesis

that the slope of the regression line is zero. The strongest structural predictor (not

including host-to-host bridging) for the mechanical outcome is in bold. (-) indicates an

inverse relationship; only shown for significant correlations. * all autografts had

callus bridging from host to host.

78

To account for other variables that contribute to the biomechanical properties

of the grafts, multivariate correlations between micro-CT parameters and torsional

properties were investigated as described in Sections 2.3.5 and 3.2.4. When included

as an independent variable, the Union Ratio was a significant, predictive variable that

increased the regression coefficients for rigidity and strength of 6 and 9 week

autografts and allografts as a group.

To determine the union ratio’s ability to improve the correlation between

structural measures and mechanics, multivariable regression was performed twice,

once without the Union Ratio, and again with the Union Ratio. Without Union Ratio,

BVGraft PMIMax and PMIMin were selected to correlate with TUlt yielding an Adjusted

R2 = 0.50 (Figure 3.3-2A) and PMIMax and PMIMin were selected to correlate with TR

yielding an Adjusted R2 = 0.31 (Figure 3.3-2C). Including Union Ratio in the

independent variables to select from resulted in significantly higher correlation

coefficients. The ultimate torque correlated significantly with the combination of

Union Ratio, BVGraft, PMIMin and the interaction terms Union Ratio×BVGraft and

Union Ratio×PMIMin (Adjusted R2 = 0.67, Figure 3.3-2B). The torsional rigidity

correlated significantly with Union Ratio, BVGraft, BVCallus, PMIMax, PMIMin, and the

interaction terms Union Ratio×PMIMax, and Union Ratio×PMIMin (Adjusted R2 =

0.57, Figure 3.3-2D).

79

(N×m

m)

(N×m

m)

(N×m

m2 )

(N×m

m2 )

Figure 3.3-2: Multivariable linear regression analysis of allografts and

autografts.

Multivariable linear regression analysis of geometric micro-CT-based parameters

including bone volume (BV), polar moment of inertia (PMI), and Union Ratio (UR).

The variable selection by regression analysis was performed without UR (A and C),

and with UR (B and D) for the combined set of autografts and allografts. Adjusted R2

and the variables that were selected to generate the optimum regression equation

are given on each graph with their coefficients and standard error indicated. Each

variable or group of variables was significant at the p < 0.05 level, as indicated by *.

80

Variables without * were part of a group of variables that significantly contributed to

the regression as a group. These groups were made of the interacting term, and the

individual terms that make of the interacting term. Upon inclusion of UR, UR was

selected as a significant contributor either by itself, or in combination with other

measures. This improved the correlation’s Adjusted R2 by 0.16 for TUlt (A and B) and

by 0.26 for TR (C and D). All regressions were significant (p < 0.01).

When allografts were analyzed separately, and before the Union Ratio was

derived, the multivariate regression analysis used BVCallus, PMIAve and PMIMax for an

Adjusted R2=0.72. With the Union Ratio available, the regression selection chose

UR, PMIMin, BVGraft, and UR×BVGraft (the interaction term) in the regression with the

ultimate torque, increasing the Adjusted R2 to 0.80 (p<0.05) (Figure 3.3-3B). The

correlation with the torsional rigidity of allografts significantly improved with the

addition of the Union Ratio from an Adjusted R2 from 0.74 to 0.89 (p<0.05) with the

combination of the Union Ratio, BVGraft, PMIMax, PMIMin and the interaction terms

with the Union Ratio: UR×BVGraft, UR×PMIMax, UR×PMIMin (Figure 3.3-3 C & D).

81

Adj.R² = 0.89

0

1000

0 1

Pred

icte

d To

rsio

nal R

igid

ity

Measured Torsional Rigidity

A B

C D

Intercept 2045 ± 538 *UR -8396 ± 3777 *BVGraft -3550 ± 841 *PMIMax 240.7 ± 110 *PMIMin 2468 ± 588 *UR×BVGraft 19741± 5430 *UR×PMIMax -3223 ± 1134 *UR×PMIMin -8593 ± 3441 *

000

Adj.R² = 0.80

0

30

0 3

Pred

icte

d U

ltim

ate

Torq

ue

Measured Ultimate Torque0

Adj.R² = 0.72

0

30

0 30

Pred

icte

d U

ltim

ate

Torq

ue

Measured Ultimate Torque

6 wk Allo9 wk Allo

Adj.R² = 0.74

0

1000

0 1000

Pred

icte

d To

rsio

nal R

igid

ity

Measured Torsional Rigidity

Intercept 27.48 ±7.81 *UR -94.48 ±62.35BVGraft -34.84 ±8.75 *PMIMin 25.08 ±6.27 *UR×BVGraft 133.6 ±60.78 *

Intercept 4.73 ±3.05BVCallus 46.50 ±11.6 *PMIAve 12.09 ±4.44 *PMIMax -15.23 ±2.87 *

Intercept -16.01 ±156BVCallus 1518 ±608 *PMIMax -401.9 ±84.7 *PMIMin 966.6 ±275 *

(N×m

m)

(N×m

m)

(N×m

m2 )

(N×m

m2 )

Figure 3.3-3: Multivariable linear regression analysis of allografts

Multivariable linear regression analysis of geometric micro-CT-based parameters

including bone volume (BV), polar moment of inertia (PMI), and Union Ratio (UR) for

allografts only. The variable selection by regression analysis was performed without

UR (A and C), and with including UR (B and D) for allografts only. Adjusted R2 and

the variables that were selected to generate the optimum regression equation are

given on each graph with their coefficients and standard error indicated. Each

variable or group of variables was significant at the p < 0.05 level, as indicated by *.

82

Variables without * were part of a group of variables that significantly contributed to

the regression as a group. These groups were made up of the interaction term and

the individual terms of the interacting term. Upon inclusion of the UR, it was selected

as a significant contributor either by itself, or in combination with other measures.

This improved the correlation’s Adjusted R2 from 0.72 to 0.80 for TUlt (A and B) and

from 0.74 to 0.89 for TR (C and D). All regressions were significant (p < 0.01).

To determine the ability of time-after-surgery as a predictor of strength and

rigidity as a factor in the regressions the correlation of time by itself was calculated

and stepwise selection was performed with time as a co-variate. By itself, time was

generally the single best predictor of strength and rigidity for the subsets of allografts-

only, autografts-only, and allografts and autografts together (Table 3.3-2). In the

multivariate analysis for TUlt, time was chosen as a significant parameter along with

BVCallus, BVGraft, and PMIMin as grouped terms including the interaction with time

(Adj. R2 = 0.85, p < 0.0001) (Details not shown). Similarly, the multivariate

regression for TR determined that time, PMIMin, and BVGraft and the interaction

time×BVGraft was the best combination of predictors (Adj. R2 = 0.82, p < 0.0001)

3.4 DISCUSSION

Despite the high incidence of bone fractures and the clinical development of

safe and effective anabolic/osteogenic therapies for bone healing (i.e. teriparatide,

BMP-2), the absence of a non-invasive outcome measure of biomechanical healing of

fractured bone continues to limit our ability to define non-unions and evaluate new

83

therapies for unmet clinical needs. In Chapter 2 the establishment of correlations of

micro-CT parameters with torsional properties in the mouse femoral graft model was

attempted, but we found that we could only predict 50% of the biomechanical

properties of the mouse grafted femurs. This poor correlation is largely explained by

the fact that none of the established micro CT parameters are capable of quantifying

the extent of cortical bone union between the graft and the host, which intuitively

should be directly related to strength of the bone. Therefore, a novel algorithm to

quantitatively estimate the union between graft and host bone based on micro-CT data

was developed and validated. We then used a combination of this and other structural

outcome measures to define regression equations would correlate bone strength and

stiffness for a spectrum of treatments which included two graft types and 2 time

points in order to objectively evaluate bone graft healing. These results successfully

highlighted the differences in healing due to graft type, as well as the changes in

union and osseointegration patterns over time. Furthermore, one-to-one correlations

demonstrated that the Union Ratio was a significant predictive variable of the

biomechanical properties of the devitalized allografts, but not the live autografts.

Quantifying the Union Ratio of live autografts and devitalized allografts

corroborated previously qualitative observations regarding the biology and

biomechanics of healing in both cases (Tiyapatanaputi 2004, and Chapter 2 of this

dissertation). Histological evidence shows that devitalized allografts induce a foreign

body reaction that encases the graft in a fibrous layer initially which can be gradually

overcome with progression of the creeping callus from the host bone that typically

84

remains restricted to the graft ends (Tiyapatanaputi et al. 2004). Our results now show

that the mitigation of non-union by 9 weeks, when the callus finally penetrates the

fibrous capsule and integrates with the devitalized allograft, significantly increases

the ultimate torque and torsional rigidity (Table 3.3-1 & Table 3.3-2).

In the case of autografts, the Union Ratio does not correlate with torsional

properties, as shown in Table 3.3-2, while the allografts’ Union Ratio significantly

correlated with the torsional properties. We hypothesize that these results reflect

fundamental biological differences in the healing of live autografts and the

devitalized allografts which arise from the contribution of periosteal cells in live

autografts that are absent in devitalized allografts. We have previously shown that

autograft repair is facilitated by both endochondral bone formation at the host-graft

junction and by intramembranous bone formation along the entire length of the graft

as early as 2 weeks post-transplantation, and undergoes significant remodeling by 4

weeks(Tiyapatanaputi et al. 2004). This results in the formation of a new bone collar

that bridges the entire length of the autograft by 4 weeks, which is also apparent in

this study at 6 and 9 weeks in Figure 3.3-1. It is hypothesized that this new bone

collar begins to assume a significant share of the in vivo loading, and therefore the

autograft begins to experience significant stress-shielding and undergoes rapid and

substantial resorption (by up to 57%) by 6 weeks, thus rendering its contribution to

mechanical properties of the femur negligible. Therefore, whether or not the

remaining graft has a high degree of union to the new cortical shell plays little role in

the overall mechanical strength. In contrast, devitalized allografts completely rely on

85

endochondral bone formation initiated by the host at the host-graft cortical junction,

with no evidence of periosteal bone formation along the length of the allograft, and

no appreciable graft resorption. The result is significant callus formation that is

limited to the host-graft junction and whose union with the allograft is crucial to load

transmission and mechanical strength.

Furthermore, our multivariate correlations do not account for the complete

cortical bridging observed in 100% of the autografts at 6 and 9 weeks, which likely

makes a significant contribution to the biomechanical properties. The development of

a measure of this type of union could potentially contribute to the ability to predict

the mechanical stability of healing bone, especially in non-critical-sized repairs where

there is substantial bridging of callus directly from host-to-host.

Previously published studies have attempted to estimate fracture and graft

union using histological and stereological techniques (Lewandrowski et al. 2002) and

2D plain radiographs (Brown et al. 2004). Unfortunately, these approaches are prone

to inaccuracies as they do not account for the 3D nature of the cortical healing.

Recent reports have attempted to utilize high-resolution micro-CT imaging to

characterize fracture non-union (Gardner et al. 2007; Dickson et al. 2008). These

studies defined measures of union based on counting the number of bridged cortices

in planar sections (Gardner et al. 2007) or relied upon qualitative 3D rendering of the

fracture sites to demonstrate union or the lack thereof in response to the treatment

(Dickson et al. 2008). Therefore, this study not only reports the development of a

novel quantitative measure of union, but to the best of our knowledge it is also the

86

first to report direct correlations between the graft and host degree of union and the

biomechanical properties of the reconstructed bone, which could have important

applications in longitudinal preclinical and clinical studies of bone repair and

grafting.

3.4.1 Clinical fracture non-union case study

The Union Ratio has significant clinical implications as a novel quantitative

biometric which prompts further study in additional pre-clinical and clinical settings.

Various preclinical and clinical studies have been performed to treat bone injuries

with adjuvant therapies to enhance healing and bone formation around allografts,

enhance their incorporation and remodeling, and their biomechanical properties and

durability. The evaluation of the repair quality and osseointegration in preclinical

animal models can be accomplished by destructive biomechanical testing. However,

the evaluation of clinical patients has to date been mostly based on non-quantitative

radiographic outcomes since destructive biomechanical testing is not an option.

To demonstrate the potential clinical utility of our algorithm on CT scans of

clinical resolution, we retrospectively analyzed clinical CT images of an anonymous

patient with a prolonged non-union (>4 months) tibial fracture, which was

subsequently non-surgically treated with teriparatide. We used our custom MATLAB

software to contour the segment of bone on one side of the fracture site similarly to

contouring around the murine graft. The surface area forming union to the other side

of the fracture was then estimated by the software. After 4 months of treatment, the

87

patient had a 2.8 fold increase in the mineralized Union Area connecting the fractured

segments which underscored the functional outcome of the patient being able to

finally bear weight on the healing leg (Figure 3.4-1).

A B

E F

C

G

D

H

Figure 3.4-1: Measuring the Union Area from clinical CT data of human

patients.

Clinical x-rays and CT scan data of a patient’s fractured tibia from before and after 4

months of teriparatide therapy were obtained retrospectively from the University of

Rochester Department of Orthopaedics, in compliance with institutional review board

research exemption. The tibial non-union 4.5 months after fracture is apparent from

plain x-ray (A). The non-union was confirmed by 3D reconstruction of the patient’s

CT as evidenced by the space between the proximal (white) and distal (blue) ends of

88

the fracture (B), which yielded a union area (red) of 4.2 cm2 (C). The remarkable

effects of teriparatide on fracture healing are demonstrated by x-ray (E) and 3D CT

(F & G), and could by quantified as a 2.8 fold increase in Union Area. Panels D & G

are rotated to show the healing surface.

Every clinical fracture case will have a different geometry of the fractured

bone, so defining how much union area is needed to be considered “healed” still

needs to be investigated. Another hurdle to overcome is how to validate the results of

being “healed” since destructive mechanical testing is impossible clinically, but they

could be compared to other outcome measures such as pain, and functional evaluation

(Puzas et al. 2006) and perhaps the re-fracture rate. Eventually, when validated in

additional pre-clinical studies, the union ratio can potentially help overcome a

significant hurdle in longitudinal clinical trials by providing a quantitative CT-based

union biometric that identify patients at risk for non-union complications.

Still, in controlled pre-clinical investigations of adjuvant therapies for bone

healing, the measurement of union can be expected to identify treatments that

accelerate osseointegration.

89

Chapter 4: The use of adjuvant systemic teriparatide (PTH 1­34) treatment improves grafted femur biomechanics by increasing graft­to­host union formation  4.1 INTRODUCTION

Large structural allografts used for reconstruction of critically-sized defects,

experience failure rates of 23 - 43% (Brigman et al. 2004; Donati et al. 2005). The

three major complicating factors are non-union (27-34%), graft fracture (24-27%) and

infection (9-16%). A component of graft fracture may also be a result of delayed

union because without union the internal fixation devices such as plates, stems and

screws are relied on for load bearing which apply stress in focused regions within

bone and thus accelerating fatigue damage (Wheeler et al. 2005). Therefore rapidly

achieving union at the graft-host interface could prevent failures due to non-unions,

and potentially failures due to fatigue fracture of the graft material. Also, without

osteointegration with the host which is the first step required for graft remodeling, the

graft material remains acellular and will not heal in the event of graft fracture.

It is well documented that intermittent administration of teriparatide, an active

peptide sequence of parathyroid hormone (PTH 1-34) is a bone anabolic factor that

enhances skeletal bone mass in osteoporotic patients (Neer et al. 2001). It has been

approved by the FDA for clinical use for osteoporosis and has been shown to reduce

the incidence of fracture (Neer et al. 2001; Body et al. 2002). Additional preclinical

90

studies show that PTH is capable of accelerating the rate of fracture healing resulting

in greater strength (Chalidis et al. 2007) as well new evidence that it is effective at

reversing persistent non-unions in patients with fragility fractures (Bukata et al.

2009). PTH has also been studied with live bone grafts such as a large segmental

vascularized bone grafts in rats to study combinational effects of PTH and

bisphosphonates (Hashimoto et al. 2007) and for enhancing spinal fusion

experimentally in conjunction with morselized autograft in rabbits (Abe et al. 2007).

The mechanism of action by which therapeutic PTH enhances bone healing

has been found to affect cell populations at multiple stages of bone healing including

the mesenchymal (Nishida et al. 1994; Kaback et al. 2008), chondrogenic (Nakazawa

et al. 2005; Kakar et al. 2007) and osteogenic (Nakajima et al. 2002; Gardner et al.

2007) cell types. With intact bone, the response to PTH treatment is dominated by a

direct effect on osteoblasts by extending their life via suppression of apoptosis thus

increasing the total number of osteoblasts (Jilka et al. 1999). In healing bone, there

appear to be additional pathways by which intermittent PTH has its anabolic effects.

It has been suggested that PTH activates the preferential differentiation of progenitor

cells towards osteoblasts (Pettway et al. 2008), but does not enhance osteoblast or

precursor proliferation (Dobnig et al. 1995).

In addition to the effects on intact bone, PTH has been shown to enhance

fracture healing to an even greater extent. PTH administered to rats recovering from

a closed tibia fracture had a greater increase in strength and callus bone volume in

their fractured femur than in their intact tibia at 40 days post-fracture (Andreassen et

91

al. 1999). Nakazawa et al has shown that by 14 days post fracture chondroprogenitor

proliferation precedes the increases in bone formation (Nakazawa et al. 2005). Thus

one mechanism may be through formation of a larger cartilage anlage prior to callus

mineralization. Similar findings were recapitulated by Kakar et al. with increased

cartilage area in PTH treated fractures at day 5 and 10, after which they found that

chondrocyte hypertrophy markers were also upregulated (Kakar et al. 2007). See

Appendix B for a table summarizing a thorough review of the literature related to

PTH for fracture healing.

We have shown that upon mechanical torsional testing of allografted mouse

femurs the weakest location after 6 weeks was at the graft-host-interface, as all the

femurs failed there (Table 2.4-2). Allograft strength is also still in great deficit at 6

weeks. The femurs with autografts implanted for 6 weeks, and allografts implanted

for 9 weeks were stronger and stiffer than 6 week allografts and the fractures location

after mechanical testing occurred within the bone of the graft and the host suggesting

that the weakest point was not exclusively at the graft-host-interface due to greater

union. In order to develop a means of quantitatively assessing the degree of union,

we then devised an algorithm to quantify this site of weakness in bone grafts that we

termed the Union Ratio. The Union Ratio is a measure of the surface area of boney

callus formation onto the surface of the graft from micro-CT images. It has been

established that the Union Ratio correlates highly with the strength and stiffness of

grafted femurs and explained over 50% of the variation in mechanical properties of

allografted femurs (Table 3.3-2).

92

The objective in this study was to determine whether graft-to-host union and

bone mechanics are improved by intermittent systemic PTH treatment at 6 weeks

after allograft implantation. We hypothesize that systemic intermittent PTH

treatment will improve the bone volume and mineral content of host callus formation

around the allograft, and will overcome the delay in graft-host union by 6 weeks.

These morphological changes of the callus will result in the recuperation of

mechanical strength and rigidity of the grafted femurs. We also use the parameters of

bone graft healing that were established earlier in this dissertation to develop a means

of explaining the variation in biomechanical strength and stiffness with non-invasive

micro-CT imaging.

4.2 METHODS

4.2.1 Experimental Model

Four millimeter long bone allografts were harvested from donor mice, and

were processed to generate clean, aseptic, devitalized bone allografts for

implantation. All soft tissue was removed, the grafts were trimmed to 4mm in length

using a diamond-sintered cutoff saw, they were bathed in 70% ethanol for 3 hours,

rinsed three times with sterile saline and frozen to -80oC for 1 week. These bone

grafts were implanted into intecalary defects in the femur of other mice and secured

in place using a 0.35mm diameter stainless steel intramedullary pin as shown in

Figure 4.2-1 and as preiviously described (Reynolds et al. 2007). One week after

surgery, daily injections of 40µg/kg hrPTH (1-34) (Lilly, Inc., Indianapolis, IN) were

93

initiated in half of the mice, while the others received injections of saline control.

Weekly x-rays were taken to monitor progression (Faxitron X-Ray LLC, Wheeling,

IL). Further procedural details for each experimental study are described in the

sections that follow.

Saline group: Daily Saline Injections

Weeks1 2 3 4 5 6

PTH group: Daily PTH Injections (40 ug/kg)

• Histology & Vascular Imaging; n=4

• MicroCT & Biomechanical Testing; n=14

• Histology & Vascular Imaging; n=4

Figure 4.2-1: Experimental Design

4.2.2 Biomechanical Study

One cohort of the study groups used 14 mice from each treatment (PTH and

control) for imaging and mechanical material testing. These were sacrificed 6 weeks

after surgery. Each femur was harvested by disarticulating the hip and knee and

removing the intramedullary stainless-steel pin. Specimens were moistened with

94

saline and frozen at -20oC until thawed for micro-CT imaging and torsion testing.

Specimens were scanned at 12.5 µm isotropic resolution using the Scanco VivaCT 40

(Scanco Medical AG, Bassersdorf, Switzerland). From these 3D images, the graft

and callus bone volumes (BVGraft, BVCallus) were measured by manual segmentation,

followed by standardized thresholding at a grayscale corresponding to 750

mgHA/cm3 based on a phantom of known HA concentrations (Nazarian et al. 2008).

The cross-sectional polar moment of inertias (PMI) were computed for each slice

throughout the grafted region and the maximum, minimum and average PMI

(PMIMax, PMIMin, PMIAve) were investigated to determine their contribution to the

biomechanics of the grafted femurs.

The Union Ratio, as described in Chapter 3 and (Reynolds et al. 2008), was

also calculated. The Union Ratio measures the graft surface area upon which

mineralized callus has formed. In brief, if the voxels adjacent to the graft surface are

boney callus, the area of that region of the graft is measured and normalized to the

total graft surface area. Figure 4.3-3 depicts the bare surface of the graft in blue, with

regions of union to the callus depicted as red. Each half (proximal and distal) of the

graft is evaluated separately and the lesser ratio of union area to total graft surface

area is given as the Union Ratio for that specimen. Callus formation that bridged

from host-to-host over the graft was determined by evaluating serial axial cross

sections from micro-CT images and given a binary result. These samples were then

mechanically tested in torsion as described in Chapter 2 and (Reynolds et al. 2007).

Yield torque (TYield), ultimate torque (TUlt), torsional rigidity (TR), toughness (or

95

work to failure) and the twist at ultimate torque were determined for each specimen.

Finally, the mode of failure for each specimen was determined using an x-ray as

depicted in Figure 2.3-4.

4.2.3 Vascularization and Histological Study

A second cohort of 16 animals underwent the same surgery with sacrifice of 8

animals at 4 weeks and 8 animals at 6 weeks post-surgery to evaluate the degree of

vascularization of the graft and callus region using vascular profusion as described

previously (Duvall 2004). Half of the animals were treated with PTH, and the other

half with saline.

4.2.3.1 Vascular perfusion

On the day of sacrifice, animals were injected with a fatal dose of ketamine

and xylazine and their vasculature was perfused using a syringe pump through a

needle placed into the left ventricle of the heart. The right atrium was also punctured

to allow the blood to drain out. They were first perfused with heparinized (100

units/ml) saline to prevent blood clotting, followed by 10% neutral-buffered formalin,

and lastly with lead-chromate contrast agent (Microfil 122, Flow Tech, Inc. Carver,

MA). Samples were fixed in 10% formalin overnight followed by harvest of the

femur and pin extraction. Samples were micro-CT scanned once after fixation, and

again after EDTA decalcification. Using both scans we could evaluate the

vasculature specifically within the mineralized callus. The vessel volume, thickness,

spacing and vessel number was determined.

96

4.2.3.2 Histology

After micro-CT imaging for vascular analysis, specimens were processed for

histology. Mid-sagittal sections were stained with alcian blue, hematoxylin, eosin

and orange G as previously described (Tiyapatanaputi et al. 2004). Micro-CT

images were manually resliced using NIH ImageJ software in the same plane as the

histology sections to compare the imaging modalities as shown in Figure 4.3-1.

4.2.4 Statistical Analysis

Student t-tests were used to compare differences between PTH treatment and

saline treatment for each of the micro-CT imaging measures, and biomechanical

testing results.

As described in Chapters 2 & 3 (Reynolds et al. 2007; Reynolds et al. 2008),

univariate regression analysis was used to determine the degree of correlation

between micro-CT imaging derived measures and ultimate torque, yield torque and

torsional rigidity. Multivariate linear regression analysis was used to determine

combinations of micro-CT parameters that correlated with the torsional mechanical

properties. Stepwise selection regression analysis was used to optimize the

combination of significant (p<0.05) independent variables in a linear model. This

was performed using SAS 9.1 (SAS Institute Inc., Cary, NC).

97

4.3 RESULTS

4.3.1 Bone analysis from Micro-CT imaging

Observations from Micro-CT imaging shown in Figure 4.3-1 and Figure 4.3-2

revealed that in PTH treated specimens host callus formation around the graft were

larger and packed with regions of trabecular bone. Intramedullary callus was also

present to a greater extent in PTH treated animals. There were also fewer apparent

non-unions visible in PTH treated specimens. Bridging over the graft from host-to-

host was present in 6 of 14 specimens from saline treated control animals, and 8 of 14

specimens treated with PTH.

At 6 weeks after surgery, PTH treatment significantly increased BVCallus by

93%, with a noteworthy increase in BVIntramed of 217% (Table 4.3-1). The enhanced

bone formation resulted in a significant 38% and 26% increase in PMIAve and PMIMax,

respectively. This was predominately due to an increase in cross sectional area due to

the increase in bone volume fraction of the callus, and not a change in the outer

diameter of the callus – the maximum outer radius was 1.8±0.2 mm for saline

controls and 1.7±0.3 mm for PTH treated animals. In PTH treated animals, bone

mineral density of the callus was significantly less dense by 14%, but the net callus

total mineral content was still significantly greater by 67% because the bone volume

fraction within the callus was 52% greater. The graft bone volume was not different

between treatment groups at 6 weeks suggesting that there was no increase in graft

resorption with PTH treatment. This was verified with histology which revealed no

difference in the resorption spaces on the graft surface area. The Union Ratio, a

98

measure of the relative surface area upon which callus bone has formed, was

significantly 76% greater (p < 0.01) (Figure 4.3-3, Table 4.3-1).

99

0.5 mm

* *

* *

* *

* *

0.2 mm

*P

IM

*P

IM

*P

IM

*

IM

0.5 mm

P

6wk

Sal

ine

4wk

PTH

6wk

PTH

BA C

ED F

HG I

KJ L

4wk

Sal

ine

Figure 4.3-1: Sagittal cross sections of grafted femurs.

Sagittal micro-CT images (A, D, G, J) of the proximal graft-host interface correspond

with the Hematoxylin/Eosin and Orange G histology images in the middle and right

100

columns. Figures A – F are from animals 4 weeks after surgery while G – L were 6

weeks after surgery. 40µg/kg of PTH(1-34) was administered daily in specimens of

figures D – F and J – L while the others received saline. Specimens in figures A – C

and G – L were also used for volumetric vascular analysis by µCT and are thus

perfused with lead-chromate contrast agent which appears white on µCT and black

in histology. Asterisks indicate the end of the graft segment, ‘IM’ indicates the

intramedullary space and ‘P’ indicates the periosteal surface. Of note is that

cartilaginous callus persisted in 4 week PTH treated specimens, PTH treatment

enhanced the ratio of bone-to-hematopoetic marrow within the callus and overcame

the fibrous the gap between callus and graft bone, thus forming callus directly on the

surface of the graft. PTH treated specimens also show enhanced intramedullary

bone formation.

101

A B C DPT

H

0.84

Salin

e

E F G H

2.21 1.37 0.22 mm3/mm

0.901.46 0.56 0.04 mm3/mm

Figure 4.3-2: Representative BV quantification from micro-CT imaging

Representative quantification of Micro-CT results for a control (A-D) and PTH treated

(E-H) specimen. Cross sectional polar moment of inertia, graft (B & F), callus (C & G)

and intramedullary callus bone (D & H) volumes were quantified for each specimen

in a region of interest that extended from the proximal axial slice containing bone

graft through the distal slice, as shown in (A,E). These regions of interest were

manually segmented and quantified for bone volume, bone mineral density and bone

mineral content at a threshold corresponding to 750 mgHA/cm3. The total depth of

penetration of callus into the intramedullary cavity from both ends was also

measured. The trabecular-like region within the shell of the exterior callus was

segmented for trabecular analysis to quantify BVF, Tb.No., Tb.Th., and Tb.Sp.

102

Table 4.3-1: Micro-CT imaging parameters of grafted femurs.

Saline PTH

PMIMin (mm4 ) 0.41 (0.11) 0.51 (0.26) PMIAve (mm4 ) 0.84 (0.14) 1.16 (0.32) ** PMIMax (mm4 ) 1.72 (0.59) 2.17 (0.46) * BVGraft (mm3 /mm) 0.84 (0.066) 0.82 (0.067) BVCallus (mm3 /mm) 0.54 (0.14) 1.04 (0.3) ** BVIntramed (mm3 /mm) 0.063 (0.042) 0.2 (0.081) ** Intramed Penetration Depth (mm) 1.73 (0.57) 2.22 (0.71) BMDCallus (mgHA/cc ) 774 (28) 667 (16) ** BMCCallus (mgHA) 1.62 (0.47) 2.7 (0.76) ** Callus trabecular BVF 0.379 (0.21) 0.576 (0.046) * Callus Tb.N. 6.38 (1.75) 13.2 (1.0) ** Callus Tb.Th. 0.0722 (0.023) 0.149 (0.198) Callus Tb.Sp. 0.182 (0.054) 0.065 (0.007) ** Union Ratio 0.129 (0.088) 0.277 (0.068) ** Mean (SD). n = 14 per treatment. * p < 0.05, ** p < 0.005

Table 4.3-2: Micro-CT imaging parameters of intact contralateral femurs.

Saline Contralateral PTH Contralateral

M-L Periosteal Diameter (mm) 1.76 (0.03) 1.85 (0.08)

A-P Periosteal Diameter (mm) 1.22 (0.001) 1.30 (0.05)

Cortical Thickness (mm) 0.180 (0.006) 0.199 (0.007) *

Cortical Bone Density (mgHA/cc) 1201 (4) 1179 (21)

Cross-Sectional Area (mm3) 0.69 (0.01) 0.82 (0.05) *

Polar Moment of Inertia (mm4) 0.27 (0.01) 0.36 (0.03) * Mean (SD) N = 8 per treatment. * p < 0.05

103

UnionRatio:0.02

UnionRatio:0.31

A B C

D

E F G

H

Figure 4.3-3: Representative CT and Union Ratio images

Volume-rendered micro-CT for a control specimen (A-D) and a PTH treated

specimen (E-H). Of note are differences in the bone callus formation as indicated by

the red brackets in (B and F) and the bone volume fraction in panels C and G as well

104

as sites corresponding to the graft-host-union location indicated by the yellow

arrows. Panels D and H show the grafts’ surfaces where red indicates regions of

mineralized callus footprint onto the graft, with blue indicating otherwise. The Union

Ratio is indicated below those surfaces. Each half of the graft is evaluated

independently, and the lesser UR is used.

4.3.2 Biomechanical Testing Results

Mechanical properties of grafted femurs 6 weeks after implantation and the

contralateral intact femurs were measured in torsion and reported in Table 4.3-3. As

expected, PTH treatment improved grafted femur torsional rigidity and strength and

failed at with less twist in a more brittle-like fashion indicating the presence of boney

union, as opposed to soft callus formation. PTH treatment doubled the torsional

rigidity of grafted femurs, returning them to equivalent of intact normal femurs.

Yield Torque was also significantly 72% greater in PTH treated grafted femurs, but

Ultimate Torque was only 23% greater (not significant). Grafted femurs from saline

treated specimens did not fail until reaching a much greater the degree of twist at TUlt

than PTH treated specimens. The rate of twist at TUlt for PTH treated specimens was

only 1/3 the rotation of control grafted specimens and were similar to the intact

contralateral femurs. Work to failure (area under the curve) was not reduced in the

saline control group because of the association of low torsional rigidity with failure at

greater deformation angles, and similar ultimate torque values. Intact contralateral

femurs from the same mice did not show a significant increase in torsional mechanics

with 6 weeks of PTH treatment, which is consistent with results from a previous

105

experiment in which intact femurs from rats receiving intermittent did not achieve a

significant increase in strength until high dose PTH (100ug/kg) was given for 8 weeks

(Hashimoto et al. 2007)

Table 4.3-3: Torsional properties of grafted and contralateral femurs in mice treated

with PTH or saline as control

Sal Graft Sal Contra PTH Graft PTH Contra Tult

(N*mm) 10.7 (4.1) 19.5 (4.8) 13.2 (5.2) 22.6 (7.3)

Tyield (N*mm) 6.8 (5.5) 13.9 (5.0) † 10.5 (4.2) * 15.1 (4.5) †

TR (N*mm2/Rad) 585 (408) 1129 (362) † 1175 (311) * 1284 (205)

Twist at Tult (Rad/mm) 0.065 (0.054) 0.025 (0.006) † 0.020 (0.018) * 0.029 (0.020)

Work to TYield (Nmm*Rad/mm)

0.0508 (0.0615) 0.134 (0.090) † 0.0687(0.0348) 0.151 (0.125) †

Work to TUlt (Nmm*Rad/mm)

0.379 (0.311) 0.286 (0.115) 0.167 (0.102) 0.401 (0.244) † p < 0.05 for graft vs. contralateral. * p < 0.05 for PTH vs saline.

After torsion testing, an x-ray of the specimens was taken to determine the

mode of failure as described in (Reynolds et al. 2007). Despite the trend that PTH-

treated specimens had fewer grafted femurs failing due to simple non-union between

the graft and host, this was not found to be statistically significant using Fisher’s

Exact test. Six of the PTH-treated specimens failed in a manner that was not simply

graft-pullout from the host, while only 3 of the saline treated specimens appeared to

have had some union.

106

Table 4.3-4: Grafted femur mode of failure after torsion testing.

Pre-Union Early Union Mature Union Saline 10 1 2 PTH 8 3 3

Correlations between micro-CT parameters and torsional properties:

In order to establish associations between micro-CT derived measures and

biomechanical outcomes, univariate and multivariate linear regression analysis was

performed. The best univariate correlations for each of the mechanical outcomes

were as follows: TR vs. UR, r2 = 0.77; TYield vs. Bridging, r2 = 0.62; TUlt vs. PMIMin,

r2 = 0.57. Table 4.3-5 shows the Pearson correlation coefficients for all the micro-CT

derived measures to the mechanical outcomes.

107

Table 4.3-5: Coefficients of determination (R2) for the univariate linear regression of

structural independent variables vs. mechanical properties TR, TYield and TUlt.

TR TYield TUlt PMIAve (mm4 ) 0.092 0.023 0.097 PMIMax (mm4 ) 0.023 0.144 0.020 PMIMin (mm4 ) 0.378 * 0.424 * 0.567 * BVGraft (mm3 /mm) 0.013 0.042 0.045 BVCallus (mm3 /mm) 0.288 * 0.139 0.203 * BVIntramed (mm3 /mm) 0.314 * 0.192 * 0.161 * BMDCallus (mgHA/cc ) 0.426 * 0.178 * 0.072 BMCCallus (mgHA) 0.202 * 0.089 0.163 * Union Ratio 0.771 * 0.589 * 0.301 * Bridging 0.403 * 0.620 * 0.534 *

* indicates significance for the two-sided test of the null hypothesis that the slope of

the regression line is zero (p < 0.05). The strongest structural predictor for the

mechanical outcome is in bold.

108

R² = 0.77090

500

1000

1500

2000

2500

0 0.1 0.2 0.3 0.4

Torsiona

l Rigidity (Nmm

2 )

UnionRatio

Torsional Rigidity vs. UnionRatio 

Saline

PTH

R² = 0.58920

5

10

15

20

25

30

0 0.1 0.2 0.3 0.4

Yield Torque

 (Nmm)

UnionRatio

Yield Torque vs. UnionRatio 

Saline

PTH

R² = 0.3010

5

10

15

20

25

30

0 0.1 0.2 0.3 0.4

Ultim

ate Torque

 (Nmm)

UnionRatio

UltimateTorque vs. UnionRatio 

Saline

PTH

R² = 0.3957

0

0.1

0.2

0.3

0 0.1 0.2 0.3 0.4

Rate of Twist a

t TUlt(Rad

/mm)

UnionRatio

Twist vs. UnionRatio 

Saline

PTH

BA

C

E

D

0 0.1 0.2 0.3 0.4UnionRatio

Mode of  Failure vs. UnionRatio 

Saline

PTH

Non-Union

Pre-Union

Mature Union

Figure 4.3-4: Linear regressions between mechanical properties and Union

Ratio.

109

The UR was found to correlate highly with TR (r2= 0.77), Tyield (r2=0.59) , TUlt (r2=0.30)

and inversely with Twist (r2=0.40). Horizontal dotted lines in A-D represent the range

of data obtained from the normal contralateral femurs in placebo-treated animals.

4.3.3 Callus Vascularization Results

Blood vessel analysis was performed using micro-CT imaging after vascular

perfusion with a contrast enhancing polymer. Quantification of the vessels within the

callus region, as shown in Figure 4.3-5 shows that there was 74% and 88% more

vessel volume in saline treated specimens at 4 and 6 weeks respectively (not

significant), which was mainly due to an increase in blood vessel diameter (55%

greater at 4 weeks p = 0.05, 78% greater at 6 weeks, p = 0.001).

110

* *

0

0.02

0.04

0.06

0.08

0.1

0.12

4 weeks 6 weeks

Vessel Thickness (mm)

0

0.2

0.4

0.6

0.8

1

4 weeks 6 weeks

Callus Vessel Volume (mm3)

0

0.5

1

1.5

2

2.5

4 weeks 6 weeks

Vessel number (1/mm)

6wks PTH6wks Saline4wks PTH4wks Saline

Saline

PTH

Figure 4.3-5: Vascularization of callus in Saline and PTH treated animals.

Representative vascular analysis via micro-CT imaging of contrast-enhancing

vascular profusion agent within the callus after decalcification. Total vascular

volume, vessel diameter and vessel number are plotted as mean ± SD; n = 4 per

group. * significance between treatment (p < 0.05).

In both PTH and saline treated animals we observed an interesting

phenomenon that a major blood vessel formed down the in the intramedullary canal

of the dead graft visible on micro-CT images of vascular-perfused specimens. This is

remarkable because despite there being vasculature within the graft, there is little or

no sign of revitalization of any other tissue associated by any other cell types. There

is neither bone nor hematopoietic marrow in the graft marrow cavity at 4 or 6 weeks

in control yet there was a single branch of the femoral artery perforating the host bone

shell and passing through the marrow cavity. This is interesting because it would

111

mean there is potential for revitalization of the graft from the interior as well as the

periphery.

6 wk PTH6 wk Saline

4 wk PTH4 wk Saline

Figure 4.3-6: Maximum intensity projections of vascular perfusion imaging.

Maximum intensity projections of micro-CT scans of intercalary allografts in mouse

femurs with vascular profusion contrast agent. Apparent in each image are

intramedullary blood vessels inside the graft which by 6 weeks span the entire space

from host to host (Arrows).

112

AdjR² = 0.840

2000

0 2000

Pred

icted TR

 (N*m

m2 )

Measured TR (N*mm2)

Measured vs. Predicted TR

NoPTHPTH

AdjR² = 0.610

25

0 25

Pred

icted T U

lt(Nmm)

Measured  TUlt (Nmm)

Measured vs. Predicted  TUlt

NoPTHPTH

AdjR² = 0.710

20

0 20

Pred

icted T Y

ield(N*m

m)

Measured TYield (N*mm)

Measured vs. Predicted Yield Torque 

NoPTHPTH

Ultimate Torque

Parameter Intercept PMIMin UnionRatio

Coefficient 2.407 14.93 12.75

Standard Error 1.582 3.135 5.734

Individual p < 0.1411 0.0001 0.04

Yield Torque

Parameter Intercept Bridging UnionRatio

Coefficient 2.030 5.662 20.11

Standard Error 1.181 1.243 5.676

Individual p < 0.1 0.0001 0.002

Torsional Rigidity

Parameter Intercept UnionRatio PMIMin

Coefficient -100.8 3151 722.5

Standard Error 97.89 354.9 194.0

Individual p < 0.31 0.0001 0.0011

Figure 4.3-7: Multivariable linear regression results.

Stepwise regression analysis was used for variable selection of micro-CT-derived

geometric properties such as segmented bone volumes, max, min or average PMIs,

Union Ratio, BMD, and host-to-host bridging, resulting in the correlations between

113

micro-CT parameters and torsional ultimate strength, yield strength and rigidity

shown here. This analysis yields correlation equations that could be used to predict

functional mechanical outcomes so the coefficients of the measures are given in the

tables.

4.4 DISCUSSION

Bone allografts are prone to high complication rates that can cause morbidity

and require interventions such as revision surgery or amputation. The major

complications after graft implantation are graft-to-host nonunion, fracture and

infection (Brigman et al. 2004; Donati et al. 2005). Graft fracture could be due to

either single loading events (trauma) or by fatigue failure. Overcoming these

complications by means of physical modification of bone grafts such as surface

decalcification, surface coating or perforation, and application of adjuvant treatments

such as BMPs, stem cell coengraftment and gene therapies has been a major topic of

study, but none of them have been clinically adopted as the gold standard for care,

while allograft failure still remains a concern.

Intermittent teriparatide (PTH 1-34), the only systemic anabolic bone factor,

has been used since before its FDA approval in 2002 for treatment of osteoporosis

(FDA 2002). PTH has an even more striking effect on enhancing bone fracture

healing. Over 40 days of 200ug/kg/day PTH treatment in rats, the percent increase in

bone mineral content of the fracture callus relative to untreated controls was 108%

while the intact contralateral femur diaphysis only enhances by BMC by 18.7% with

114

PTH treatment (Andreassen 1999). In this study we investigated the use of

intermittent teriparatide for standard cortical allografts to determine if the reported

anabolic effect in fracture healing also improves allograft bone incorporation. This

original work shows the anabolic effect of intermittently administered PTH (1-34)

effectively stimulates callus formation around bone allografts. This robust callus

overcomes the delay in radiographic non-union by 6 weeks which is an improvement

over normal allografts and plays a significant role in improving biomechanical

strength and stiffness.

Cartilage formation was increased with PTH treatment, which persisted

through 4 weeks after surgery, which was similar to results published by Nakazawa in

which PTH enhanced cartilage formation at the site of bone injury (Figure 4.3-1)

(Nakazawa et al. 2005). This extensive cartilage would then undergo ossification via

chondrocyte hypertrophy, thus suggesting that one mode of PTH's effect, which

resulted in greater bone volume at 6 weeks, was due to enhanced cartilaginous callus

formation.

There was also an improved graft-to-host union ratio apparent on micro-CT in

animals treated with PTH which meant that PTH treatment was able to overcome the

formation of the fibrous layer that forms around implanted bone grafts

(Tiyapatanaputi et al. 2004), (Stevenson et al. 1997) and hinders their incorporation

with the host callus. The mechanism by which PTH achieved greater union is not

specifically known. It could be due to simply a filling of the space between graft and

callus, or it could be that PTH accelerates the recruitment of stem cells to the wound

115

site before the fibrous response can mount. It could also be that PTH drives

mesenchymal stem cell differentiation toward the chondroblastic and/or osteoblastic

phenotypes, rather than the fibroblastic (Nakajima et al. 2002; Nakazawa et al. 2005;

Rickard et al. 2006). Studying the underlying basis for this to determine what local

factors could be responsible for reversing non-unions could be investigated in future

research. This will be discussed further in Chapter 5.

Side-by-side comparison of histological and radiographic imaging of the

union of the callus to allografts indicates that union, in this case, is attributable to

callus bone forming directly adjacent to the graft, but not necessarily integrating with

that graft tissue via remodeling of the graft initiated from the callus (Figure 4.3-1).

To some degree this distinguishes radiographic union from histological union. Here,

we found that radiographic union was sufficient to improve bone biomechanics.

As previously published, we found a preferential enhancement of total bone

mineral content of the callus at 6 weeks compared to the enhancement of the systemic

skeletal bone mineral content. The ratio of callus BMCPTH:BMCSaline was 1.67

whereas the contralateral intact femur’s diaphyseal BMCPTH:BMCSaline = 1.17. Zhou

et al. noted differences in the change in mineralization level of the axial skeleton

relative to the appendicular skeleton with intermittent PTH treatment, finding that the

axial skeleton (vertebral bodies) had greater increases in BMD (Zhou et al. 2003).

They suggested that this is due to the greater trabecular number in the axial skeleton

which therefore has more surface area upon which osteoblasts can form bone. In

addition to the increased cartilaginous callus volume early, the increased surface area

116

during callus formation may be another reason why there is greater improvement in

callus bone volume than in the intact contralateral diaphysis. PTH treatment appears

to affect multiple stages of bone healing, with enhancement of cartilage early which

turns into callus, as well as a greater bone formation rate of the trabecular-like woven

bone of the callus.

Figure 4.3-3A-D shows an obvious proximal non union in the saline control,

which corresponds to a Union Ratio of 0.02, which is interpreted as only 2% of that

half of the graft is in contact with callus, whereas the PTH-treated specimen in Figure

4.3-3H with UR = 0.31 has at least 31% of either half of the graft upon which callus

had formed. Across all samples (see Table 4.3-1), the Union Ratio was greater by 2.8

fold in PTH treated animals. Attaining a level of union that corresponds to a

recuperation of the mechanical properties of intact femurs can be identified in the

plots of Figure 4.3-4. The intersection of the trend line with the dashed line

representing the range of values for normal femurs indicates a threshold at which

union could be considered sufficient. With the various mechanical outcome

measures, this UR ranged from 0.12 to 0.23 with a mean intersection of 0.18. It can

be inferred that achieving that level of union in this mouse model would mean

returning the risk of limb fracture to “normal”. Interestingly, this did not correspond

to a dramatic shift in the location of failure of these femurs (Table 4.3-4). There was

a trend that fewer femurs failed in a non-union mode, but this trend was not

significant. This suggests that PTH induced robust callus formation adjacent to the

graft, but it may not have integrated with the graft.

117

The result of the improvements in the callus by PTH treatment resulted in

stronger graft biomechanics. In fact, it was found that these structural parameters

correlate highly with the biomechanical strength and stiffness determined by torsion

testing. Univariate regression analysis revealed that as expected, many of the

structural bone geometry and density measures correlated strongly with mechanical

outcome measures. According to their coefficient of determination (R2) the best of

these were the Union Ratio, the host-to-host bridging and PMIMin (Table 4.3-5).

Multivariable regression analysis of all the imaging-derived parameters showed that

by pairing combinations of those best three predictors the correlation significantly

improves, adding 0.14 – 0.17 to the adjusted coefficients of determination (Adj. R2)

(Figure 4.3-7).

The revitalization of the intramedullary canal of the graft with bone is a novel

observation and points to another means of revitalizing graft tissue from the inside

out. Intramedullary bone formation has not been observed in any of our studies

aimed at enhancing mouse allograft bone healing when adjuvant treatments were

locally delivered on the periosteal surface of the graft. These studies included the use

of rAAV-caAlk2 (Koefoed et al. 2005), combination rAAV-Vegf and rAAV-RankL

(Ito et al. 2005), rAAV-BMP2 (unpublished) and co-engraftment of C9 stem cells

(Xie et al. 2007). This increase in BVIntramed was mostly due to increased bone

volume fraction and a small increase in the depth of penetration of boney callus into

the grafts from both ends. Intramedullary penetration depth was 28% greater in PTH

treated animals (2.2 ± 0.7 mm in grafts from PTH-treated animals, 1.7 ± 0.6mm in

118

grafts from saline-treated animals; p = 0.12) but intramedullary callus that was

present was densely layered with bone. As on the exterior surface of allografts, the

cell types present in the intramedullary canal of allografts from saline treated mice

were predominantly fibroblastic, but with PTH treatment the composition of the

intramedullary space at the ends of the graft were predominantly osteoblastic cells.

Another novel observation was that a major intramedullary blood vessel was

visible on micro-CT images of animals with vascular contrast agent. This was

identified as a penetrating branch of the femoral artery that re-bridged, over time,

from host to host inside the graft. At 4 weeks the vessel was visible, and by 6 weeks

it had bridged the length of the graft in 3 of 4 saline treated specimens, and 4 of 4

PTH treated specimens. From histology it is apparent that the graft intramedullary

space is largely void of healing callus or hematopoetic marrow, and instead only

sparse fibroblasts and adipocytes. It may have been assumed that there was no

nutrient supply to this intramedullary space thus graft regeneration should focus on

the periosteal surface, but knowing now that vascular invasion of the graft is present

suggests that adjuvant local therapeutics in the intramedullary space should not be

ignored as a means of graft revitalization. Studying the revascularization of bone

grafts using the intramedullary canal as an indicator of graft revitalization may or

may not be appropriate as we found intramedullary vascularization with little or no

graft revitalization in both PTH and Saline treated specimens. Resolving the

vasculature within the graft material itself may be a better indicator. This deserves

further investigation.

119

Although patients receiving large structural allografts after removing bone

neoplasms would be restricted from PTH treatment due to an assumed increased risk

of cancer (Vahle et al. 2002), there are many other uses of bone grafts such as for

trauma, joint revision arthroplasty, dental implants, oral surgery, and spinal fusion in

which tumors are not involved for which PTH could be utilized without the risk of

exacerbating tumor recurrence.

Although a study of morselized autograft for spinal fusion shows early

enhancement of osteoclast-related genes and an increase in osteoclast number (Abe et

al. 2007), and another study of fracture healing in rats showed a significant increase

in OC# per fracture callus area (Komatsubara et al. 2005), we found that PTH

treatment for 6 weeks did not stimulate osteoclastic graft resorption, and thus there

was also no increase in revitalization of the graft tissue through remodeling.

Contrarily, other studies of fracture calluses show no increase in osteoclast number

per bone callus surface area beyond 1 week post-fracture (Nakajima et al. 2002;

Alkhiary et al. 2005; Manabe et al. 2007). These discrepancies may be due to the

method of counting osteoclasts, whether it is normalized to the cross-sectional callus

area, or the perimeter of the mineralized callus surface. To know whether

osteoclastic resorption of the graft can be stimulated via continuous elevation of PTH

(or by some other controlled means) should be investigated. PTH could also be used

in conjunction with other therapies as a control mechanism to maintain the highest

level of graft integrity. The toolbox of systemically administered therapies would

120

then include intermittent PTH for callus formation, short-term bisphosphonates to

inhibit osteoclastic resorption and perhaps continuous PTH to stimulate it.

The timing of PTH initiation after injury may be an important control

parameter for engineering a rehabilitation regime. We initiated daily saline or PTH

injections one week after initial surgery to allow normal hematoma formation to

complete before handling the animals daily. Pettway et al recently performed a study

of bone tissue engineering to investigate the effect of PTH on bone marrow stromal

cells (BMSC) at different points of differentiation (Pettway et al. 2008). They

showed that the effect of intermittent PTH was greatest on BMSCs that had been

implanted for one week suggesting that PTH may have its greatest effect on

stimulating pre-osteoblast proliferation.

Based on our results we find that the anabolic effect of PTH can significantly

improve callus formation from the host around bone grafts and for the first time show

a potential solution to improving bone graft-to-host union which would significantly

alleviate problems with graft non-unions. This could reduce the need for additional

surgical interventions in patients with non-stable constructs. If proven to be safe and

effective, there is potential for the use of PTH immediately after surgery to reduce the

recovery time and prevent other graft complications.

121

Chapter 5: Conclusions and Future Research Suggestions  5.1 REVIEW OF RESEARCH

Bone grafts are commonly used for skeletal reconstruction for a variety of

indications but experience complications. These complications include infection,

incomplete union, microcrack accumulation due to a lack of to intracortical repair,

fatigue failure, and should fracture occur within the graft material it is highly unlikely

to heal. Clinicians and researchers are aware of these limitations, but have few

alternatives that are superior. There is a paucity of tools for forcasting bone graft fate

after implantation. Generating tools that can quantitatively evaluate bone healing

could help identify patients at risk for complications, as well evaluate bone graft

alternatives and adjuvant therapies in pre-clinical models to establish the best

candidates. The research presented here establishes a method for performing

mechanical and structural quantification of bone graft fate after implantation in a

mouse model, devises a new tool to directly measure how osteoconductive a graft

surface is, and validates that delivering a bone anabolic factor stimulates bone callus

formation and reverses the inhibition of union formation.

Most notable from this work was that this model of bone allografting in mice

results in a delay in union up to 6 weeks which is recovered by 9 weeks, and results in

significant improvement in rigidty and stiffness. There appeared to be a trend of

decreasing strength after 9 weeks in allografts which was most likely due to a

122

combination of graft resorption and fatigue within the graft material. In a side

project, a method for visualizing microdamage was developed to attempt to

investigate whether microdamage accumulation was apparent in allografts between 9

and 12 weeks (see Section 5.2, below). At 6 weeks in autografts, there was increased

degree of union of callus bone onto the graft which was more dispersed along the

length of the graft since healing was enhanced by the periosteal callus formation.

Autograft strength similarly tended to decline over time until 18 months, but to a

lesser extent, possibly due to stress shielding of the bone by the intramedullary pin.

This conclusion is supported by clinical and research experience in which stress

shielding due to internal fixation devices can cause bone atrophy.

As in other animal and clinical studies, treatment with teriparatide rapidly

improved callus formation which improved radiographic allograft union and resulted

in greater stability of grafted femurs. This, along with other corroborating research

suggests that PTH may be effective for improving bone healing in many situations.

Multiple studies in fractures (see Appendix B) and additionally in spinal fusion using

morselized autograft (Abe et al. 2007) and live vascular fibular autografts (Hashimoto

et al. 2007) have all shown improvements using intermittent PTH therapy. PTH’s

effects are more pronounced in healing bone than they are in normal skeletal tissue,

suggesting that the cascade of events using intermittent PTH could be somewhat

different in healing bone than in mature bone.

123

5.2 MICRODAMAGE STUDY

The reduction in torsional strength and rigidity of bone allografts in our mouse

model after 9 weeks may be due in part to the accumulation of microdamage within

the graft material. To evaluate the amount of microdamage within the graft material a

method was developed to visualize microdamage histologically.

Normal mouse femurs were taken fresh and underwent cyclic loading using an

Instron Dynamite 8841 (Instron, Norwood MA) materials testing device in 3-point

bending for up to 1000 cycles to induce microdamage. These femurs were used as

controls to optimize the labeling and embedding protocols and confirm that

microdamage labeling was identifiable. For the experimental specimens, bone

allografts (4mm long) were implanted into the mid diaphysis of 10 mice which were

sacrificed at 9 and 12 weeks post-surgery (n=5 per time point). The intramedullary

pins were extracted from femurs, and the bones were fixed using 70% ethanol

overnight, labeled under vacuum in 1.5x10-4 molar calcein in 70% ethanol for 4

hours. Processing for plastic PMMA embedding involved rinsing and dehyrdrating in

a gradient of ethanol over days, cleared using xylazine, and infiltrated with PMMA

polymer over 2 days, and finally embedded in PMMA and polymerized over days

using the benzoyl peroxide decomposition technique. This polymerization is initiated

by adding benzoyl peroxide to the PMMA solution and incrementally raising the

temperature by 1°C every 12 hours from room temperature until polymerization is

completed. PMMA blocks are then sectioned with a IsoMet slow speed saw using a

0.4mm thick diamond wafering blade (Buehler Lake Bluff, IL). These sections are

124

then polished using incrementally finer cloths (600 grit, 1500 grit and 4000 grit) and

imaged using epifluorescence or confocal microscopy.

The protocol described above was the result of many iterations using a limited

number of samples, and therefore, many of the specimens were not suitable to be

properly imaged. Figure 5.2-1 Microdamage in mouse cortical bone Figure 5.2-1

shows representative histology from normal femurs showing the successful labeling

of intracortical surfaces (osteocytes) as well as the existence of microcracks in a

fatigued specimen. The limited samplings at 9 and 12 weeks did not reveal obvious

microdamage accumulation in this model. Instead, large resorption spaces were most

apparent within the graft tissue, especially in 12 week allografts. This is consistent

with the reduced graft bone volume (20%) measured from CT quantification (Figure

2.4-1). Due to the limited number of specimens, and infrequent identification of

microcracks, quantification was not pursued. However, future efforts should

investigate this phenomenon in a systematic study that evaluates allograft healing

over time and in response to therapeutic interventions to enhance repair.

125

A

B

Figure 5.2-1 Microdamage in mouse cortical bone

Bulk staining of cortical mouse bone with calcein fluorescence labeling reveals

intracortical surfaces such as osteocytes lacunae within normal bone (A), and bone

that was subjected to cyclic loading (B) using epifluorescence at 20x magnification.

126

5.3 RATIONALE FOR FUTURE RESEARCH DIRECTIONS

5.3.1 Further invesitigation of Union Metrics

5.3.1.1 Other Clinical Applications

The software tools for measuring the union onto a graft surface as presented in

Chapter 3 are flexible and capable of measuring other 3D surfaces, generating a shell

around them, and quantifying the amount of callus which penetrates that shell. This

has been applied to a variety of clinical fractures cases in pilot studies. Clinical

fractures can occur in any part of the skeleton, and have infinite conformations of the

fracture shape. Therefore it is not clear how much of the fracture surface needs to be

fused to the other fracture surface to achieve adequate union. This question will be

discussed more in 5.3.1.2. These pilot investigations showed that in patients with

non-union fractures, the fractured segment could successfully be identified in 3D

from standard clinical CT imaging. Fractures which present as non unions were

predominantly in the cervical spine, and in distal tibia and fibula. As these patients

were osteoporotic and had been on conservative treatment for at least 4 months

without successful consolidation of their fractures, they were placed on Forteo

(teriparatide) (Bukata et al. 2009). Through functional assessment, pain evaluation,

and radiographic imaging, approximately 93% of these patients’ persistent non-

unions were successfully reversed, achieving union. In 8 cases, the degree of union

was measured using the system described in Chapter 3 which corroborated the finding

of successful union area between the fractured segments by showing measurable

increases in the union surface area of the fracture in the majority of these patients.

127

Unfortunately, each fracture surface is unique and so the amount of surface area

necessary to achieve union is unknown, and so more work needs to be done to make

the measure of union meaningful from a diagnostic perspective. One way in which

this measure could be useful in this setting is to measure the rate of change in union

area over time. This could be achieved by longitudinal imaging with at least three

time points measured: two images captured at different times before initialization of

treatment and a third is captured after some time with treatment. To evaluate a

potential therapy, one could measure the change in the rate of union area before and

after treatment and compare that change in rate to a placebo treatment. Evaluating

the change in the relative change of the union area would eliminate variation. Further

study of this concept should be pursued to validate the Union Ratio as a meaningful

indicator of a therapy’s efficacy.

Another straightforward application for the Union Ratio technique lies in

evaluating prosthetic implant equipment. Dental implants, for instance, are typically

threaded metal sockets that are screwed into the mandible or maxilla and covered for

a number of weeks to allow for osseointegration onto their surface. It has been found

that osseointegration is relatively successful in the anterior mandible (97%), but in the

posterior maxilla the success rate is lower (77%) (Tolstunov 2007). The predominant

reason for this is lower bone quality in the posterior maxilla which limits the amount

of bone growth onto the implant. Evaluating the amount of bone contact with the

implant would be one way of identifying patients at risk for implant loosening and

could be evaluated using CT imaging. One technique for sampling a titanium

128

implant’s osseointegration was done by radial point counting on 2D axial slices

(Kuroda et al. 2004) where radial lines passing from the center of the implant were

investigated. The number of lines radiating from the implant that crossed into bone

first, instead of marrow space, counted toward that implant’s level of

osteointegration. They found that synthetic coating of the implant with apatite

improved radiographic osseointegration and torsional strength. Another study used

multilevel thresholding to evaluate the percent of bone pixels relative to the total

number of pixels adjacent to the implant to measure the %osseointegration (%OI)

(Gabet et al. 2006). They found that high dose teriparatide (75ug/kg) treatment

improved %OI by and that 25 and 75 mg/kg teriparatide improved pullout strength of

the implant. The advantage of studying prosthetic implants is that the surface area

can be known. The mechanical result of changes in union onto the implant can be

determined using finite element modeling and preclinical models to validate findings,

then further studies can be based simply on the changes in union.

Automation of the surface generation for defining the boundary of bone and

prosthetic implants is an area which could be improved for the Union Ratio

algorithm. Currently, images are semi-manually contoured (See Figure 3.2-1) which

can be time consuming and subject to user interpretation. Automatic mesh generation

could be accomplished by scanning bone grafts prior to or immediately after

implantation. This would show the known geometry of the graft material. A 3D shell

should be easily generated around this pure representation of the graft which could

later be registered to the remaining graft in the follow-up scan. This would be very

129

possible for prosthetic implants which often have an attenuation coefficient that is

different than bone. Then implants could be used to generate their own shell.

5.3.1.2 How much union is needed? When is a fracture “healed”?

Knowing what percentage of the surface of a bone segment is in contact with

surrounding callus is an appropriate measure for comparing the osteoconduction

between samples, and between treatments. We have shown in our model how union

ratio and mechanics are related, but determining when a specific specimen has

achieved adequate union to consider it fully healed remains complicated. For our

grafts, “union” could be the level needed to achieve the mechanical properties of the

range of normal contralateral limbs (see Figure 4.3-4). Further work must be

performed to understand how much union area needs to be achieved in a diverse array

of fracture types.

There are several conceivable methods to start understanding how much union

area is needed. One method would be to use normal healthy bone as a template. For

example, transferring the shell generated around our segment of interest (the graft) to

an intact femur, the surface area of connection between the virtual graft and the host

would be the transverse cross sectional area of that segment (since the osteotomies we

make are transverse). This can set a basis to compare the area of union of actual

grafts. In this example, the femoral cross sectional area normalized to the surface

area of a half a graft (as our Union Ratio measure does) is approximately 0.06. The

data shown in Figure 4.3-4 do not support this as being an adequate level of union.

130

There are two possible reasons for this discrepancy: the distribution of the union

surface on the graft is different, and there is a surface-bonding component that is

unaccounted for. To address the question about how the distribution of the union

affects the strength, a virtual graft model could be generated from the CT images, and

then the surface area of union could be modified to change the pattern of distribution

on the surface. Applying a set of different loading conditions (torque, bending about

the M-L Axis, bending about the A-P axis) to each of the models a finite element

analysis program could be produced. The magnitude of the maximum stress in each

model would then illustrate the risk of failure within that model. The patterns of

bonding could vary from a single continuous union area at the end (as the transverse

cross sectional area mentioned above) to a random distribution over the surface of the

graft as was observed in autografts at 6 and 9 weeks.

Understanding what the adhesion properties of the newly formed callus are on

the graft would also need to be accomplished. As shown in Figure 4.3-1, the

radiographic union of callus in the teriparatide treated animals involved bone

formation onto the surface of the graft. Since no additional remodeling of the graft

was stimulated, there was probably not true integration with the graft. I.e. the mineral

crystals were not continuous from the graft to the host. Accounting for the bond

strength would be important to reconcile that perfect continuous bone has a “Union

Ratio” of 0.6, while grafts with a union ratio of 2 or 3 times that have similar strength

and still fail at the graft-host interface (Table 4.3-2, Table 4.3-3, and Table 4.3-4).

One way of measuring this adhesion would be to implant cortical bone graft dowels

131

into a host bone of an animal. Micro CT imaging for union area measurement and

pullout testing could be performed to understand the adhesive properties of the graft-

host-interface.

5.3.1.3 An alternative measure of overall connectivity

Host to host bridging was a phenomenon found in autografts by 6 weeks, in

allografts at later time points, and in many of the allografts in the PTH study at 6

weeks. Simply noting if bridging was present was able to explain 40 – 62% of the

variation in mechanical properties of grafted femurs (Table 4.3-5). Quantifying the

degree of bridging may be able to enhance our understanding of the torsional

properties. Two considerations related to bridging are how thick is the bridge, and

what is its spatial relationship to the bending or torquing axes? One way of

quantifying bridging would be to measure the minimum thickness of the bridge.

Because the geometry is complicated, this thinnest plane could be at any angle

possible (not just the axial plane) across the specimen. For example, the digital

phantom in Figure 3.2-2 has examples of both transverse and oblique planes of graft-

to-host bridging. So it is somewhat impossible to quantify the minimum cross section

of bridging in all planes. During part of this dissertation, an alternative was

investigated. In this technique traces (lines) are drawn from one plane of interest to

another through the material (bone pixels in a CT image for instance). A series of

requirements for the drawing of this line are followed to achieve the measurement of

the thickness of a bridge between the two planes. The trace is drawn to hug the

132

surface of the bone with the goal of making its way toward the "goal" plane. Only

one trace may pass through a bone pixel (subsequent traces will hug the boundary

between the bone and the previous traces). The first trace (which is teal in Figure

5.3-2) starts on one edge of the start plane (the bottom left corner), and each

subsequent trace starts next to the previous trace. The line is drawn to hug the edge

of the bone area and will fill up a region of bone until it finds an outlet and can reach

the Goal plane. When a trace reaches a point where it can no longer go towards the

goal plane, by following the bottom or right surface of the bone, it will return to a

previous location within the trace to continue searching.

In the case of the teal trace, there were no connections on the bottom half of

the image, and so the bone was filled with that trace, until it reached a bridge to the

goal plane on the upper half of the image. This finally occurred at the place above the

black void in the host callus along which the trace makes a right-turn in the lower left

corner of the image. The next trace (in fucia) starts at the next available location

along the start plane, and ends up following alongside the first trace until it reaches

the goal plane. Three more traces do the same until a constriction indicated by the

red arrow is filled with 5 traces total. A subsequent trace in green can find no way of

reaching the goal plane, and thus fills the region Counting the number of traces that

successfully bridge one plane to another effectively quantifies the width of the

constriction between. Figure 5.3-1 gives a representation of this technique in 2D.

Transitioning this to 3D is more complicated and has not yet been solved, but is

possible.

133

Start Plane Goal Plane

Figure 5.3-1: 2D Flow connectivity example.

134

On a 2D cross sectional image of a grafted femur, tracings were performed

automatically by a program written in Matlab. Tracings started on the bottom of the

start plane with the goal plane as the end point. Each tracing is colorized differently

for visualization purposes.. Five tracings successfully reached their target end point.

One constriction site is indicated by the red arrow.

As mentioned before, the location of this minimum cross section or

constriction site relative to the bending or torsional axes would play a role in its

contribution to the overall strength. Identifying the constriction site is not straight

forward in 3D, and would have to be studied further. After the constriction site is

identified, its contribution to the structure could be studied.

5.3.1.4 Large animal models of grafting and the application of union ratio

The union ratio has been investigated using CT images from a published study

of a single clinical allograft in which a high resolution investigational cone-beam CT

scanner was used (Ehrhart et al. 2008). Using these images posed multiple challenges

that would need to be address to pursue further clinical studies. Metal implants,

particularly steel alloys, have very high attenuation coefficients which generate image

artifacts obscuring details near these implants. Artifact suppression algorithms have

been developed for minimizing their effect, but are not yet standard. These artifacts

contribute to the uncertainty of the measurement of the union surface between a graft

and the host. None the less, union measures were obtained for this case, as shown in

Figure 5.3-2. It is obvious that parts of the graft will not be analyzed, but in regions

135

that are free from artifact, the union area could be measured. Selection of plates

made of less-radiopaque materials such as titanium instead of stainless steel would

generate less striking artifacts. Also a sample region of the surface of the graft that is

free from artifact could be selected for union area quantification, and monitored over

time in a longitudinal study.

6 Month FollowupWhole Area:

9300mm2

Connected Area:1500 mm2

BaselineWhole Area:

8500 mm2

Connected Area:1700 mm2

Plate and screw artifact Intramedullary stem

A B

C

Figure 5.3-2: Case study: Union area of a proximal tibia allograft

The proximal tibia from a clinical allograft was imaged with high resolution cone

beam CT. The sagittal and axial cross secions through the tibia are shown in panels

A and B. Apparent union area was measured around the graft and shown in red. Of

136

note are significant areas of artifact and locations where the intramedullary stem

passed through the shell drawn around the graft.

The research of this dissertation addresses one of the major complications of

allograft failure: non-union. The second major complication requiring further

investigation is the accumulation of microdamage leading to fatigue failure.

Identifying the risk factors that cause it would be paramount to minimizing its

occurrence. I propose one possible interaction between these complications, which is

that non unions contribute to microdamage accumulation due to reliance on implant

hardware to bear the load. Studying a larger animal model of bone grafting would

allow for the application of locking plates and screws that interact similarly to the

clinical setting. The first step would be to validate the union measurement in this

model despite implant hardware. Next, it would need to be validated that the model

induces microdamage in the graft material in a timely manner, and if necessary

increasing the animal’s activity or even using externally applied loading to induce

microdamage. Finally, determining if a relationship exists between how quickly a

specimen establishes union and the rate at which it develops microdamage or fatigue

failure would be instrumental in advocating augmentation of the graft-host junction to

prevent the majority of allograft failures. Pilot work for this research could initially

be done in cadaveric bone, using surgical reconstruction techniques, followed by

simulation of various degrees of graft-host union with cement, for instance.

137

5.3.2 Radiation exposure and justification for clinical CT imaging.

Computed tomographic radiography exposes the subject to potentially harmful

ionizing radiation that should not be undertaken without an evaluation of the risks and

benefits to a patient. Recent advances in radiography have continued to minimize the

exposure levels while continuing to achieve adequate resolution. Alongside these

advances are improvements in diagnostic capabilities through qualitative and

quantitative image analysis. The risk-to-health need of a patient needs to be

considered on multiple levels to include of the risk from a single exposure as well as a

patient's cumulative exposure over time. The age of the patient, and any existing

conditions also contribute in that patient's risk.

The measure of the radiation exposure are typically reported in Gray (Gy) or

Sievert (Sv) and is the joules of radiation exposure per kilogram. For x-rays and

gamma rays, 1Gy = 1Sv = 100 Rad. As a point of reference, approximate radiation

exposures for posterior-anterior chest x-ray is 0.01 mSv, lateral chest x-ray is 0.15

msV and an adult abdominal CT gives 10 mSv (Brenner et al. 2007). Lattanzi et al

found that a CT scan of the pelvis and femur for pre-operative planning of hip

replacement surgery results in approximately 5 mSv (range 2 - 7 mSv) (Lattanzi et al.

2004). They also quote that baseline whole body exposure is approximately 2mSv

per year (Farr et al. 1997).

A study of microCT dose exposure in rats was performed to investigate the

effects of longitudinal exposure on normal healthy rat bone. The Scanco Viva CT 40

scanner was used to image the rat hind limb at 15 µm resolution and resulted in an

138

exposure of somewhere between 422 and 987 mGy (Brouwers et al. 2007). This CT

protocol achieves very high resolution (15 µm) which is comparable to the 10.5 –

12.5 micron resolution in the much smaller animal used in this study, as the mouse

body is 1/10th the weight of rats. Brouwers et al. found no significant effect on

trabecular geometrical values over time, nor any effect on the vitality of bone marrow

cells due to scans performed weekly for 7 weeks. Another study of the radiation

exposure to mice using a lower resolution (200 µm) to image the torso found

exposure levels between 80 and 160 mGy (Taschereau et al. 2006).

The actual adverse effects of radiation exposure caused by CT scanning has

not been fully studied. Instead, estimates of the risk of cancer due to radiation

exposure are mainly based on studies of atomic-bomb survivors and those employed

in the nuclear industry (Brenner et al. 2007). Those studies show that radiation

exposure in the 5 to 150 mSv significantly increases the risk of developing cancer.

Radiation exposure to youths is more risky than to adults and increase the potential

for cancer because they youths are inherently more sensitive to radiation, and also

have a greater number of years to develop radiation-induced cancer. They estimate

that the overall risk of death due to cancer from a "typical" series of 2 or 3 CT scans

of the head or the chest for an adult is between 0.01% and 0.02% and for a child is

between 0.02% and 0.1%. Although the individual's risk is small, they estimate that

1.5 – 2% of all cancers may be attributable to current CT usage, and should be

considered a general public health risk.

139

The method by which a CT scan is acquired can vary the overall exposure

considerably. Cassese et al found that the radiation dose measured from 4 different

clinical CT scanners using the same high resolution technique (slice thickness =

1mm) may vary by a factor of 3 (Cassese et al. 2003) depending on the scanner. One

significant variable in radiation dose necessary to achieve a given resolution is the

detector geometry. Multidetector CT scanners can achieve greater resolution with

less total radiation exposure. There can be a 100% difference in the radiation

exposure for the same scan resolution using more refined detector configurations

(Dalrymple et al. 2007). Even more promising for high resolution imaging with

lower radiation exposure is the advent of cone beam CT (CBCT) imaging. Cone

beam CT imaging has been developed and investigated particularly in oncology field

for diagnostic purposes, as in breast cancer (Chen et al. 2002), as well as for radiation

oncology treatment planning and where multiple imaging sessions are used to ensure

proper patient positioning each time a patient undergoes fractionated external beam

radiation treatments. Studies comparing CBCT to the more standard multidetector

CT imaging systems indicate that radiation exposure can be 35 to 49% lower for head

and body scans, respectively (Kim et al. 2008). Cone beam CT imaging optimized

for detection of breast tumors (soft tissue, low contrast, high penetrance specimen)

can achieve spatial resolution of 0.36 mm with a delivered dose of 336 to 235 mRad

(2.35 – 3.36 mSv) depending on the size of the breast (Chen et al. 2002). This is

relatively high resolution with low dose, compared to the clinical CT scanners

140

described above, but differs due to total volume being imaged, and material

composition.

A longitudinal clinical study evaluating massive osteoarticular allografts with

CT imaging detected fractures earlier in the graft material with CT imaging than

standard radiographs, and thus supports CT imaging to evaluate the condition of a

patient's the bone allograft (Mattila et al. 1995). A clinical study comparing the

qualitative assessment of fracture union using CT and planar radiography in the

appedicular skeleton found CT imaging is not significantly more sensitive in

evaluating the time-to-union in most examples, but CT imaging was necessary in

11% of cases for which the assessment of healing fractures could not be determined

from standard plain x-rays due to positioning or internal hardware (Grigoryan et al.

2003). Fractures of the axial skeleton can be particularly difficult to evaluate from

planar x-rays. These studies indicate that the evaluation of clinical allografts with CT

imaging could identify complications due to fracture or resorption and would be

recommended for improved prognosis. Although the risk inherent with ionizing

radiation exists, improvements in the imaging resolution along with reduction of total

dose are being made will allow for quantitative assessment through CT image

analysis and will be beneficial to patient care.

5.3.3 Teriparatide Therapy

Our investigation of PTH treatment for bone graft healing showed that the

induction of an osteoblastic response filled the space between the outer callus shell

141

and the graft surface with bone, forming a tightly woven network through which load

could be transferred from the host to the graft. This was not seen in saline treated

allografts nor in autografts (where the cortical shell was maintained while the graft

was resorbed). The rampant bone formation reduced the volume of hematopoietic

marrow in the callus region, a soft tissue which would not contribute to graft stability.

Reduction in vascular volume was concomitantly measurable, mostly due to a

reduction in vessel diameter, rather than vessel number or distribution. The decrease

in hematopoietic tissue and vascular volume in the high bone density callus, is

contrary to our expectations. PTH promotes osteoblast-coupled hematopoiesis

(Whitfield 2005). The anabolic effect of PTH can be attenuated with an angiogenesis

inhibitor (Rhee et al. 2007). Osteogenesis is coupled with angiogenesis (Gerber et al.

2000; Wang et al. 2007) and active osteoblasts express vascular endothelial growth

factor (VEGF) (Deckers et al. 2000). But our finding and another recent report

(Prisby ASBMR Montreal 2008) indicate that osteogenesis can be stimulated without

an increase in angiogenesis. Future work could evaluate whether combining an

angiogenesis treatment via rAAV-Vegf (as shown in Ito 2005) in combination with

systemic PTH treatment would enhance bone formation more markedly.

Recently, a concern was raised about a possible decrease in strength of

implant fixation after termination of PTH treatment in which rats were given

intermittent PTH for two weeks. After PTH withdrawl they were treated with either

saline or bisphosphonate for three weeks (Johansson et al. 2008). They found that the

effect of terminating the administration of PTH after this initial healing phase

142

returned the pull-out strength of the screw to the same level as screws from untreated

animals. They also found the loss of strength after PTH withdrawal could be

prevented with the use of a bisphosphonate. Therefore, further studies would need to

be conducted to elucidate the effect on grafted bone strength after suspension of

anabolic PTH administration and whether bisphosphonates would be recommended

upon termination of PTH treatment.

Torsional resistance of grafted femurs was recovered with the use of PTH

treatment for 6 weeks, but did not stimulate graft remodeling and there was no

increase in osteoclast activity on the surface of the graft observed. Dead allograft

tissue is subject to fatigue damage accumulation for which targeted remodeling would

be an attractive means of reversing. Conversely, over-zealous graft resorption is a

potential risk of using an osteoclast stimulating therapy such as rAAV-RANKL (Ito et

al. 2005). Fortunately we now know that systemic PTH treatment can promote callus

formation, and could be used as a control agent to stimulate bone formation when

necessary.

Hypocalcemia and in hyperparathyroidism cause elevated serum PTH levels.

This causes osteoclastic resorption from the skeleton. Could sustained PTH treatment

be used to stimulate osteoclastic resorption of graft bone? This could be achieved via

local expression delivered via gene therapy, or by systemic elevation using an

osmotic pump. This could serve to be a non invasive control mechanism for inducing

osteoclastic resorption of the graft material after successful callus formation.

143

Many massive bone graft recipients are receiving allografts to replace a bone

neoplasm. It is thought that PTH carries a risk of stimulating cancer, thus precluding

treatment of these patients. At very high doses, PTH has been described as

carcinogenic in rats, but only when given at dose-levels that are 50 or 150ug/kg body

weight which is 27 and 66 times the exposure used clinically, but not at the level of

4.7 times the clinical dose (Jolette et al. 2006). Due to these studies, there is a black-

box limitation on the use of Forteo in patients with cancer to prevent adverse events.

There have been no clinical reports of increased tumorigenic incidence due to

teriparatide (Harper et al. 2007) and a long term study in non-human primates at

doses closer to clinical doses has not been found to cause cancer (Vahle et al. 2008).

This observed mature ovariectomized monkeys for 4.5 years after initialization of

daily teriparatide treatment 5 μg/kg body weight that was given for 18 months, there

were no incidents of neoplasia (Vahle et al. 2008). Further research could be

conducted to address whether PTH stimulates proliferation of existing neoplasms. In

vitro work with osteosarcoma and other cancer cell lines exposed to PTH, and in vivo

studies in mice could address the potential for risk.

144

6 References 

Abe, Y., M. Takahata, et al. (2007). "Enhancement of graft bone healing by intermittent administration of human parathyroid hormone (1-34) in a rat spinal arthrodesis model." Bone 41(5): 775-85.

Albert, A., T. Leemrijse, et al. (2006). "Are bone autografts still necessary in 2006? A three-year retrospective study of bone grafting." Acta Orthop Belg 72(6): 734-40.

Alkhiary, Y. M., L. C. Gerstenfeld, et al. (2005). "Enhancement of experimental fracture-healing by systemic administration of recombinant human parathyroid hormone (PTH 1-34)." J Bone Joint Surg Am 87(4): 731-41.

Andreassen, T. T., C. Ejersted, et al. (1999). "Intermittent parathyroid hormone (1-34) treatment increases callus formation and mechanical strength of healing rat fractures." J Bone Miner Res 14(6): 960-8.

Aro, H., E. Eerola, et al. (1982). "Osteolysis after rigid fixation. The possible role of periosteal neural mechanoreceptors in bone remodeling." Clin Orthop Relat Res(166): 292-300.

Aslan, H., Y. Zilberman, et al. (2006). "Nucleofection-based ex vivo nonviral gene delivery to human stem cells as a platform for tissue regeneration." Tissue Eng 12(4): 877-89.

Attias, N. and R. W. Lindsey (2006). "Case reports: management of large segmental tibial defects using a cylindrical mesh cage." Clin Orthop Relat Res 450: 259-66.

Baer, D. G., T. J. Walters, et al. (2004). "Bone and Soft Tissue Trauma Research at the USAISR." RTO HFM Symposium RTO-MP-HFM-109: P35-1 - P35-10.

Bentolila, V., T. M. Boyce, et al. (1998). "Intracortical remodeling in adult rat long bones after fatigue loading." Bone 23(3): 275-81.

Berrey, B. H., Jr., C. F. Lord, et al. (1990). "Fractures of allografts. Frequency, treatment, and end-results." J Bone Joint Surg Am 72(6): 825-33.

Blokhuis, T. J., F. C. den Boer, et al. (2000). "Evaluation of strength of healing fractures with dual energy Xray absorptiometry." Clin Orthop Relat Res(380): 260-8.

Body, J. J., G. A. Gaich, et al. (2002). "A randomized double-blind trial to compare the efficacy of teriparatide [recombinant human parathyroid hormone (1-34)] with alendronate in postmenopausal women with osteoporosis." J Clin Endocrinol Metab 87(10): 4528-35.

Bonadio, J., E. Smiley, et al. (1999). "Localized, direct plasmid gene delivery in vivo: prolonged therapy results in reproducible tissue regeneration." Nat Med 5(7): 753-9.

145

Boutroy, S., M. L. Bouxsein, et al. (2005). "In vivo assessment of trabecular bone microarchitecture by high-resolution peripheral quantitative computed tomography." J Clin Endocrinol Metab 90(12): 6508-15.

Boyce, T., J. Edwards, et al. (1999). "Allograft bone. The influence of processing on safety and performance." Orthop Clin North Am 30(4): 571-81.

Brenner, D. J. and E. J. Hall (2007). "Computed tomography--an increasing source of radiation exposure." N Engl J Med 357(22): 2277-84.

Bresler, F., P. Simon, et al. (1998). "Digital image analysis of bone allograft union in sheep." Acta Orthop Scand 69(2): 181-3.

Brigman, B. E., F. J. Hornicek, et al. (2004). "Allografts about the Knee in Young Patients with High-Grade Sarcoma." Clin Orthop Relat Res(421): 232-9.

Brodt, M. D., C. B. Ellis, et al. (1999). "Growing C57Bl/6 mice increase whole bone mechanical properties by increasing geometric and material properties." J Bone Miner Res 14(12): 2159-66.

Brouwers, J. E., B. van Rietbergen, et al. (2007). "No effects of in vivo micro-CT radiation on structural parameters and bone marrow cells in proximal tibia of wistar rats detected after eight weekly scans." J Orthop Res 25(10): 1325-32.

Brown, K. M., M. M. Saunders, et al. (2004). "Effect of COX-2-specific inhibition on fracture-healing in the rat femur." J Bone Joint Surg Am 86-A(1): 116-23.

Broz, J. J., S. J. Simske, et al. (1993). "Effects of rehydration state on the flexural properties of whole mouse long bones." J Biomech Eng 115(4A): 447-9.

Bruder, S. P., D. J. Fink, et al. (1994). "Mesenchymal stem cells in bone development, bone repair, and skeletal regeneration therapy." J Cell Biochem 56(3): 283-94.

Bukata, S. V., L. A. Kaback, et al. (2009). "1-34 PTH at Physiologic Doses in Humans Shows Promise as a Helpful Adjuvant in Difficult to Heal Fractures: An Observational Cohort of 145 Patients." ORS Meeting Las Vegas.

Burchardt, H. (1983). "The biology of bone graft repair." Clin Orthop Relat Res(174): 28-42.

Burr, D. B., R. B. Martin, et al. (1985). "Bone remodeling in response to in vivo fatigue microdamage." J Biomech 18(3): 189-200.

Burr, D. B. and T. Stafford (1990). "Validity of the bulk-staining technique to separate artifactual from in vivo bone microdamage." Clin Orthop Relat Res(260): 305-8.

Burwell, R. G. (1963). "Studies in the transplantation of bone. V. The capacity of fresh and treated homografts of bone to evoke transplantation immunity." J Bone Joint Surg Br 45-B: 386-401.

Camacho, N. P., C. M. Rimnac, et al. (1995). "Effect of abnormal mineralization on the mechanical behavior of X-linked hypophosphatemic mice femora." Bone 17(3): 271-8.

Cassese, J. A., A. S. Brody, et al. (2003). "Utility of simple radiation dose measurements in the evaluation of different CT scanners used for high-resolution CT." J Thorac Imaging 18(4): 242-5.

146

Chalidis, B., C. Tzioupis, et al. (2007). "Enhancement of fracture healing with parathyroid hormone: preclinical studies and potential clinical applications." Expert Opin Investig Drugs 16(4): 441-9.

Chen, B. and R. Ning (2002). "Cone-beam volume CT breast imaging: feasibility study." Med Phys 29(5): 755-70.

Chen, J. B., Y. Yu, et al. (2007). "BMP-7 and CBFA1 in allograft bone in vivo bone formation and the influence of gamma-irradiation." J Biomed Mater Res A 80(2): 435-43.

Chen, Y., K. D. Luk, et al. (2003). "Gene therapy for new bone formation using adeno-associated viral bone morphogenetic protein-2 vectors." Gene Ther 10(16): 1345-53.

Cloward, R. B. (1980). "Gas-sterilized cadaver bone grafts for spinal fusion operations. A simplified bone bank." Spine 5(1): 4-10.

Cook, S. D., R. L. Barrack, et al. (2000). "The Otto Aufranc Award. Strut allograft healing to the femur with recombinant human osteogenic protein-1." Clin Orthop Relat Res(381): 47-57.

Cruess, R. L. and J. Dumont (1975). "Fracture healing." Can J Surg 18(5): 403-13. Dalrymple, N. C., S. R. Prasad, et al. (2007). "Price of isotropy in multidetector CT."

Radiographics 27(1): 49-62. Damron, T. A., W. G. Ward, et al. (2007). "Osteosarcoma, chondrosarcoma, and

Ewing's sarcoma: National Cancer Data Base Report." Clin Orthop Relat Res 459: 40-7.

Day, J. S., M. Ding, et al. (2000). "Parallel plate model for trabecular bone exhibits volume fraction-dependent bias." Bone 27(5): 715-20.

Deal, C. and J. Gideon (2003). "Recombinant human PTH 1-34 (Forteo): an anabolic drug for osteoporosis." Cleve Clin J Med 70(7): 585-6, 589-90, 592-4 passim.

Deckers, M. M., M. Karperien, et al. (2000). "Expression of vascular endothelial growth factors and their receptors during osteoblast differentiation." Endocrinology 141(5): 1667-74.

Defense, D. o. (2008). "Global War on Terrorism Casualities by Reason " Retrieved 11/29/08, from http://siadapp.dmdc.osd.mil/personnel/CASUALTY/gwot_reason.pdf.

Delloye, C., O. Cornu, et al. (2007). "Bone allografts: What they can offer and what they cannot." J Bone Joint Surg Br 89(5): 574-9.

Delloye, C., L. Coutelier, et al. (1986). "Canine cortical bone autograft remodeling in two simultaneous skeletal sites." Arch Orthop Trauma Surg 105(2): 79-99.

Delloye, C., P. Simon, et al. (2002). "Perforations of cortical bone allografts improve their incorporation." Clin Orthop Relat Res(396): 240-7.

den Boer, F. C., P. Patka, et al. (1999). "New segmental long bone defect model in sheep: quantitative analysis of healing with dual energy x-ray absorptiometry." J Orthop Res 17(5): 654-60.

Desandes, E. (2007). "Survival from adolescent cancer." Cancer Treat Rev 33(7): 609-15.

147

Dickson, G. R., C. Geddis, et al. (2008). "Microcomputed tomography imaging in a rat model of delayed union/non-union fracture." J Orthop Res 26(5): 729-36.

Dobnig, H. and R. T. Turner (1995). "Evidence that intermittent treatment with parathyroid hormone increases bone formation in adult rats by activation of bone lining cells." Endocrinology 136(8): 3632-8.

Doi, K., S. Kawai, et al. (1988). "Anterior cervical fusion using the free vascularized fibular graft." Spine 13(11): 1239-44.

Donati, D., C. Di Bella, et al. (2005). "The use of massive bone allografts in bone tumour surgery of the limb." Current Orthopaedics 19: 393-399.

Dubuc, F. L. and M. R. Urist (1967). "The accessibility of the bone induction principle in surface-decalcified bone implants." Clin Orthop Relat Res 55: 217-23.

Ehrhart, N., S. Kraft, et al. (2008). "Quantification of massive allograft healing with dynamic contrast enhanced-MRI and cone beam-CT: a pilot study." Clin Orthop Relat Res 466(8): 1897-904.

Einhorn, T. A. (1998). "The cell and molecular biology of fracture healing." Clin Orthop Relat Res(355 Suppl): S7-21.

Enneking, W. F. and D. A. Campanacci (2001). "Retrieved human allografts : a clinicopathological study." J Bone Joint Surg Am 83-A(7): 971-86.

Enneking, W. F. and E. R. Mindell (1991). "Observations on massive retrieved human allografts." J Bone Joint Surg Am 73(8): 1123-42.

Esiashvili, N., M. Goodman, et al. (2008). "Changes in incidence and survival of Ewing sarcoma patients over the past 3 decades: Surveillance Epidemiology and End Results data." J Pediatr Hematol Oncol 30(6): 425-30.

Fang, J., Y. Y. Zhu, et al. (1996). "Stimulation of new bone formation by direct transfer of osteogenic plasmid genes." Proc Natl Acad Sci U S A 93(12): 5753-8.

Farr, R. F. and P. J. Allisy-Roberts (1997). Physics for medical imaging. London, Saunders Company.

FDA (2002). "FDA APPROVES TERIPARATIDE TO TREAT OSTEOPOROSIS." FDA Talk Paper(T02-49).

Flick, L. M., J. M. Weaver, et al. (2003). "Effects of receptor activator of NFkappaB (RANK) signaling blockade on fracture healing." J Orthop Res 21(4): 676-84.

Forwood, M. R. and C. H. Turner (1995). "Skeletal adaptations to mechanical usage: results from tibial loading studies in rats." Bone 17(4 Suppl): 197S-205S.

Friedlaender, G. E. (1991). "Bone allografts: the biological consequences of immunological events." J Bone Joint Surg Am 73(8): 1119-22.

Friedlaender, G. E., K. W. Sell, et al. (1978). "Bone allograft antigenicity in an experimental model and in man." Acta Med Pol 19(1-2): 197-205.

Frost, H. M. (1960). "Presence of microscopic cracks in vivo in bone." H. Ford Hosp. Med. Bull. 8: 25–35.

Fukuroku, J., N. Inoue, et al. (2007). "Extracortical bone-bridging fixation with use of cortical allograft and recombinant human osteogenic protein-1." J Bone Joint Surg Am 89(7): 1486-96.

148

Fuller, K., J. M. Owens, et al. (1998). "Induction of osteoclast formation by parathyroid hormone depends on an action on stromal cells." J Endocrinol 158(3): 341-50.

Gabet, Y., R. Muller, et al. (2006). "Parathyroid hormone 1-34 enhances titanium implant anchorage in low-density trabecular bone: a correlative micro-computed tomographic and biomechanical analysis." Bone 39(2): 276-82.

Gafni, Y., G. Pelled, et al. (2004). "Gene therapy platform for bone regeneration using an exogenously regulated, AAV-2-based gene expression system." Mol Ther 9(4): 587-95.

Gardner, M. J., M. C. van der Meulen, et al. (2007). "Role of parathyroid hormone in the mechanosensitivity of fracture healing." J Orthop Res 25(11): 1474-1480.

Garrison, K. R., S. Donell, et al. (2007). "Clinical effectiveness and cost-effectiveness of bone morphogenetic proteins in the non-healing of fractures and spinal fusion: a systematic review." Health Technol Assess 11(30): 1-150, iii-iv.

Gazit, D., A. Ben-Arav, et al. (2008). "Craniofacial Bone Tissue Engineering using rAAV-BMP2-Coated Calvaria Allografts." TERMIS Conference.

Gerber, H. P. and N. Ferrara (2000). "Angiogenesis and bone growth." Trends Cardiovasc Med 10(5): 223-8.

Grigoryan, M., J. A. Lynch, et al. (2003). "Quantitative and qualitative assessment of closed fracture healing using computed tomography and conventional radiography." Acad Radiol 10(11): 1267-73.

Harper, K. D., J. H. Krege, et al. (2007). "Osteosarcoma and teriparatide?" J Bone Miner Res 22(2): 334.

Hartman, E. H., P. H. Spauwen, et al. (2002). "Donor-site complications in vascularized bone flap surgery." J Invest Surg 15(4): 185-97.

Hashimoto, T., M. Shigetomi, et al. (2007). "Sequential treatment with intermittent low-dose human parathyroid hormone (1-34) and bisphosphonate enhances large-size skeletal reconstruction by vascularized bone transplantation." Calcif Tissue Int 81(3): 232-9.

Hidaka, C., M. E. Cunningham, et al. (2006). "Modern biologics used in orthopaedic surgery." Curr Opin Rheumatol 18(1): 74-9.

Hopp, S. G., L. E. Dahners, et al. (1989). "A study of the mechanical strength of long bone defects treated with various bone autograft substitutes: an experimental investigation in the rabbit." J Orthop Res 7(4): 579-84.

Hornicek, F. J., Jr., W. Mnaymneh, et al. (1998). "Limb salvage with osteoarticular allografts after resection of proximal tibia bone tumors." Clin Orthop Relat Res(352): 179-86.

Huff, T. W. and T. P. Sculco (2007). "Management of bone loss in revision total knee arthroplasty." J Arthroplasty 22(7 Suppl 3): 32-6.

Huja, S. S., M. S. Hasan, et al. (1999). "Development of a fluorescent light technique for evaluating microdamage in bone subjected to fatigue loading." J Biomech 32(11): 1243-9.

Huja, S. S., T. R. Katona, et al. (1999). "Microdamage adjacent to endosseous implants." Bone 25(2): 217-22.

149

Isaksson, H., I. Grongroft, et al. (2008). "Remodeling of fracture callus in mice is consistent with mechanical loading and bone remodeling theory." J Orthop Res.

Ito, H., J. J. Goater, et al. (2004). "Light-activated gene transduction of recombinant adeno-associated virus in human mesenchymal stem cells." Gene Ther 11(1): 34-41.

Ito, H., M. Koefoed, et al. (2005). "Remodeling of cortical bone allografts mediated by adherent rAAV-RANKL and VEGF gene therapy." Nat Med 11(3): 291-7.

Jane, J. A., Jr., B. A. Dunford, et al. (2002). "Ectopic osteogenesis using adenoviral bone morphogenetic protein (BMP)-4 and BMP-6 gene transfer." Mol Ther 6(4): 464-70.

Jilka, R. L., R. S. Weinstein, et al. (1999). "Increased bone formation by prevention of osteoblast apoptosis with parathyroid hormone." J Clin Invest 104(4): 439-46.

Johansson, H. R., R. Skripitz, et al. (2008). "Bisphosphonates can block the deterioration in implant fixation after withdrawal of intermittent doses of parathyroid hormone." J Bone Joint Surg Br 90(3): 400-4.

Jolette, J., C. E. Wilker, et al. (2006). "Defining a noncarcinogenic dose of recombinant human parathyroid hormone 1-84 in a 2-year study in Fischer 344 rats." Toxicol Pathol 34(7): 929-40.

Jones, A. L., R. W. Bucholz, et al. (2006). "Recombinant human BMP-2 and allograft compared with autogenous bone graft for reconstruction of diaphyseal tibial fractures with cortical defects. A randomized, controlled trial." J Bone Joint Surg Am 88(7): 1431-41.

Kaback, L. A., Y. Soung do, et al. (2008). "Teriparatide (1-34 human PTH) regulation of osterix during fracture repair." J Cell Biochem 105(1): 219-26.

Kagan, R. J. (1998). Standards for Tissue Banking. McLean, VA, American Association of Tissue Banks.

Kakar, S., T. A. Einhorn, et al. (2007). "Enhanced chondrogenesis and Wnt signaling in PTH-treated fractures." J Bone Miner Res 22(12): 1903-12.

Kakiuchi, M. and K. Ono (1987). "The relative clinical efficacy of surface-decalcified and wholly decalcified bone alloimplants." Int Orthop 11(2): 89-94.

Kakiuchi, M. and K. Ono (1996). "Preparation of bank bone using defatting, freeze-drying and sterilisation with ethylene oxide gas. Part 2. Clinical evaluation of its efficacy and safety." Int Orthop 20(3): 147-52.

Kang, J. S. and N. H. Kim (1995). "The biomechanical properties of deep freezing and freeze drying bones and their biomechanical changes after in-vivo allograft." Yonsei Med J 36(4): 332-5.

Kass, M., A. Witkin, et al. (1988). "Snakes: Active Contour Models." International Journal of Computer Vision 1(4): 321.

Kerry, R. M., B. A. Masri, et al. (1999). "The biology of bone grafting." Instr Course Lect 48: 645-52.

150

Kim, S., T. T. Yoshizumi, et al. (2008). "Comparison of Radiation Doses between Cone Beam Ct and Multi Detector Ct: Tld Measurements." Radiat Prot Dosimetry.

Knothe Tate, M. L., T. F. Ritzman, et al. (2007). "Testing of a new one-stage bone-transport surgical procedure exploiting the periosteum for the repair of long-bone defects." J Bone Joint Surg Am 89(2): 307-16.

Koefoed, M., H. Ito, et al. (2005). "Biological effects of rAAV-caAlk2 coating on structural allograft healing." Mol Ther 12(2): 212-8.

Komatsubara, S., S. Mori, et al. (2005). "Human parathyroid hormone (1-34) accelerates the fracture healing process of woven to lamellar bone replacement and new cortical shell formation in rat femora." Bone 36(4): 678-87.

Kotobuki, N., Y. Katsube, et al. (2008). "In vivo survival and osteogenic differentiation of allogeneic rat bone marrow mesenchymal stem cells (MSCs)." Cell Transplant 17(6): 705-12.

Kuroda, S., A. S. Virdi, et al. (2004). "A low-temperature biomimetic calcium phosphate surface enhances early implant fixation in a rat model." J Biomed Mater Res A 70(1): 66-73.

Laird, R. K., N. J. Pavlos, et al. (2006). "Bone allograft non-union is related to excessive osteoclastic bone resorption: a sheep model study." Histol Histopathol 21(12): 1277-85.

Lattanzi, R., F. Baruffaldi, et al. (2004). "Specialised CT scan protocols for 3-D pre-operative planning of total hip replacement." Med Eng Phys 26(3): 237-45.

Lee, K., J. D. Deeds, et al. (1994). "Parathyroid hormone induces sequential c-fos expression in bone cells in vivo: in situ localization of its receptor and c-fos messenger ribonucleic acids." Endocrinology 134(1): 441-50.

Lee, T. C., S. Mohsin, et al. (2003). "Detecting microdamage in bone." J Anat 203(2): 161-72.

Lekishvili, M. V., A. I. Snetkov, et al. (2004). "Experimental and clinical study of the demineralized bone allografts manufactured in the tissue bank of CITO." Cell Tissue Bank 5(4): 231-8.

Lewandrowski, K. U., L. Bonassar, et al. (1998). "Mechanical properties of perforated and partially demineralized bone grafts." Clin Orthop Relat Res(353): 238-46.

Lewandrowski, K. U., S. Bondre, et al. (2002). "Porous poly(propylene fumarate) foam coating of orthotopic cortical bone grafts for improved osteoconduction." Tissue Eng 8(6): 1017-27.

Lewandrowski, K. U., G. Schollmeier, et al. (2001). "Incorporation of perforated and demineralized cortical bone allografts. Part I: radiographic and histologic evaluation." Biomed Mater Eng 11(3): 197-207.

Lewandrowski, K. U., G. Schollmeier, et al. (2001). "Incorporation of perforated and demineralized cortical bone allografts. Part II: A mechanical and histologic evaluation." Biomed Mater Eng 11(3): 209-19.

151

Liu, H., D. M. Kemeny, et al. (2006). "The immunogenicity and immunomodulatory function of osteogenic cells differentiated from mesenchymal stem cells." J Immunol 176(5): 2864-71.

Locklin, R. M., S. Khosla, et al. (2003). "Mediators of the biphasic responses of bone to intermittent and continuously administered parathyroid hormone." J Cell Biochem 89(1): 180-90.

Lodder, M. C., W. F. Lems, et al. (2004). "Reproducibility of bone mineral density measurement in daily practice." Ann Rheum Dis 63(3): 285-9.

Lord, G., J. H. Marotte, et al. (1988). "Cementless revisions of failed aseptic cemented and cementless total hip arthroplasties. 284 cases." Clin Orthop Relat Res(235): 67-74.

Loty, B., J. P. Courpied, et al. (1990). "Bone allografts sterilised by irradiation. Biological properties, procurement and results of 150 massive allografts." Int Orthop 14(3): 237-42.

Loty, B., B. Tomeno, et al. (1994). "Infection in massive bone allografts sterilised by radiation." Int Orthop 18(3): 164-71.

MacDonald, B. T., D. M. Joiner, et al. (2007). "Bone mass is inversely proportional to Dkk1 levels in mice." Bone 41(3): 331-9.

Mallows, C. L. (1973). "Some Comments on Cp." Technometrics 15: 661-675. Manabe, T., S. Mori, et al. (2007). "Human parathyroid hormone (1-34) accelerates

natural fracture healing process in the femoral osteotomy model of cynomolgus monkeys." Bone 40(6): 1475-82.

Manfrini, M., D. Vanel, et al. (2004). "Imaging of vascularized fibula autograft placed inside a massive allograft in reconstruction of lower limb bone tumors." AJR Am J Roentgenol 182(4): 963-70.

Markel, M. D., R. L. Morin, et al. (1991). "Quantitative CT for the evaluation of bone healing." Calcif Tissue Int 49(6): 427-32.

Martin-Badosa, E., D. Amblard, et al. (2003). "Excised bone structures in mice: imaging at three-dimensional synchrotron radiation micro CT." Radiology 229(3): 921-8.

Mattila, K. T., J. T. Heikkila, et al. (1995). "Massive osteoarticular knee allografts: structural changes evaluated with CT." Radiology 196(3): 657-60.

McGraw, K. O. and S. P. Wong (1996). "Forming Inferences About Some Intraclass Correlation Coefficients." Psychological Methods 1(1): 30 - 46.

Meadows, G. R. (2002). "Adjunctive use of ultraporous beta-tricalcium phosphate bone void filler in spinal arthrodesis." Orthopedics 25(5 Suppl): s579-84.

Mellick, L. B., L. Milker, et al. (1999). "Childhood accidental spiral tibial (CAST) fractures." Pediatr Emerg Care 15(5): 307-9.

Mihelic, N. E. (2004). "Repair of a severely comminuted distal femur fracture using surgical-grade calcium sulfate bone graft substitute." Orthopedics 27(1 Suppl): s127-8.

Mikic, B., M. C. van der Meulen, et al. (1995). "Long bone geometry and strength in adult BMP-5 deficient mice." Bone 16(4): 445-54.

152

Muller, M., D. Mitton, et al. (2008). "Nonlinear ultrasound can detect accumulated damage in human bone." J Biomech 41(5): 1062-8.

Nakajima, A., N. Shimoji, et al. (2002). "Mechanisms for the enhancement of fracture healing in rats treated with intermittent low-dose human parathyroid hormone (1-34)." J Bone Miner Res 17(11): 2038-47.

Nakazawa, T., A. Nakajima, et al. (2005). "Effects of low-dose, intermittent treatment with recombinant human parathyroid hormone (1-34) on chondrogenesis in a model of experimental fracture healing." Bone 37(5): 711-9.

Nather, A., A. Thambyah, et al. (2004). "Biomechanical strength of deep-frozen versus lyophilized large cortical allografts." Clin Biomech (Bristol, Avon) 19(5): 526-33.

Nauta, A. J., G. Westerhuis, et al. (2006). "Donor-derived mesenchymal stem cells are immunogenic in an allogeneic host and stimulate donor graft rejection in a nonmyeloablative setting." Blood 108(6): 2114-20.

Nazarian, A., B. D. Snyder, et al. (2008). "Quantitative micro-computed tomography: a non-invasive method to assess equivalent bone mineral density." Bone 43(2): 302-11.

Neer, R. M., C. D. Arnaud, et al. (2001). "Effect of parathyroid hormone (1-34) on fractures and bone mineral density in postmenopausal women with osteoporosis." N Engl J Med 344(19): 1434-41.

Nguyen, H., D. A. Morgan, et al. (2007). "Sterilization of allograft bone: effects of gamma irradiation on allograft biology and biomechanics." Cell Tissue Bank 8(2): 93-105.

Nicholls, P. J., E. Berg, et al. (1979). "X-ray diagnosis of healing fractures in rabbits." Clin Orthop Relat Res(142): 234-6.

Niemeyer, P., A. Seckinger, et al. (2004). "[Allogenic transplantation of human mesenchymal stem cells for tissue engineering purposes: an in vitro study]." Orthopade 33(12): 1346-53.

Nishida, S., A. Yamaguchi, et al. (1994). "Increased bone formation by intermittent parathyroid hormone administration is due to the stimulation of proliferation and differentiation of osteoprogenitor cells in bone marrow." Bone 15(6): 717-23.

Nunamaker, D. M. (1998). "Experimental models of fracture repair." Clin Orthop Relat Res(355 Suppl): S56-65.

O'Brien, F. J., D. Taylor, et al. (2000). "Visualisation of three-dimensional microcracks in compact bone." J Anat 197 Pt 3: 413-20.

Okubo, Y., K. Bessho, et al. (2000). "Osteoinduction by recombinant human bone morphogenetic protein-2 at intramuscular, intermuscular, subcutaneous and intrafatty sites." Int J Oral Maxillofac Surg 29(1): 62-6.

Otsu, N. (1979). "A threshold selection method from gray-level histograms." IEEE Trans. Systems, Man, and Cybernetics 9(1): 62-66.

Owens, B. D., J. C. Wenke, et al. (2006). "Extremity trauma research in the United States army." J Am Acad Orthop Surg 14(10 Suppl): S37-40.

153

Pelker, R. R. and G. E. Friedlaender (1987). "Biomechanical aspects of bone autografts and allografts." Orthop Clin North Am 18(2): 235-9.

Pelker, R. R., G. E. Friedlaender, et al. (1983). "Biomechanical properties of bone allografts." Clin Orthop Relat Res(174): 54-7.

Pelker, R. R., J. McKay, Jr., et al. (1989). "Allograft incorporation: a biomechanical evaluation in a rat model." J Orthop Res 7(4): 585-9.

Pettway, G. J., J. A. Meganck, et al. (2008). "Parathyroid hormone mediates bone growth through the regulation of osteoblast proliferation and differentiation." Bone 42(4): 806-18.

Pike, R. L. and P. J. Boyne (1974). "Use of surface-decalcified allogeneic bone and autogenous marrow in extensive mandibular defects." J Oral Surg 32(3): 177-82.

Pluhar, G. E., P. A. Manley, et al. (2001). "The effect of recombinant human bone morphogenetic protein-2 on femoral reconstruction with an intercalary allograft in a dog model." J Orthop Res 19(2): 308-17.

Prewitt, J. M. S. (1970). Object enhancement and extraction. New York, Academic Press.

Puzas, J. E., J. Houck, et al. (2006). "Accelerated fracture healing." J Am Acad Orthop Surg 14(10 Suppl): S145-51.

Reddi, A. H. (1998). "Role of morphogenetic proteins in skeletal tissue engineering and regeneration." Nat Biotechnol 16(3): 247-52.

Rees, D. C. and F. S. Haddad (2003). "Bone transplantation." Hosp Med 64(4): 205-9.

Reynolds, D. G., C. Hock, et al. (2007). "Micro-computed tomography prediction of biomechanical strength in murine structural bone grafts." J Biomech 40(14): 3178-86.

Reynolds, D. G., S. Shaikh, et al. (2008). "Micro-CT-Based Measurement of Cortical Bone Graft-to-Host Union." J Bone Miner Res.

Rhee, Y., S. Y. Park, et al. (2007). "Angiogenesis inhibitor attenuates parathyroid hormone-induced anabolic effect." Biomed Pharmacother.

Rickard, D. J., F. L. Wang, et al. (2006). "Intermittent treatment with parathyroid hormone (PTH) as well as a non-peptide small molecule agonist of the PTH1 receptor inhibits adipocyte differentiation in human bone marrow stromal cells." Bone 39(6): 1361-72.

Sample, S. J., M. Behan, et al. (2008). "Functional adaptation to loading of a single bone is neuronally regulated and involves multiple bones." J Bone Miner Res 23(9): 1372-81.

Sandhu, H. S., H. S. Grewal, et al. (1999). "Bone grafting for spinal fusion." Orthop Clin North Am 30(4): 685-98.

Santoni, B. G., N. Ehrhart, et al. (2008). "Effects of low intensity pulsed ultrasound with and without increased cortical porosity on structural bone allograft incorporation." J Orthop Surg 3: 20.

154

Santoni, B. G., W. J. Womack, et al. (2007). "A mechanical and computational investigation on the effects of conduit orientation on the strength of massive bone allografts." Bone 41(5): 769-74.

Sautin, E. N. (1963). "[Sterilization of Bone Tissue with Gamma Rays of Cobalt-60.]." Radiobiologiia 3: 621-5.

Seeherman, H. J., M. Bouxsein, et al. (2004). "Recombinant human bone morphogenetic protein-2 delivered in an injectable calcium phosphate paste accelerates osteotomy-site healing in a nonhuman primate model." J Bone Joint Surg Am 86-A(9): 1961-72.

Seiler, J. G., 3rd and J. Johnson (2000). "Iliac crest autogenous bone grafting: donor site complications." J South Orthop Assoc 9(2): 91-7.

Sharrard, W. J. and D. H. Collins (1961). "The fate of human decalcified bone grafts." Proc R Soc Med 54: 1101-2.

Shasha, N., S. Krywulak, et al. (2003). "Long-term follow-up of fresh tibial osteochondral allografts for failed tibial plateau fractures." J Bone Joint Surg Am 85-A Suppl 2: 33-9.

Shefelbine, S. J., U. Simon, et al. (2005). "Prediction of fracture callus mechanical properties using micro-CT images and voxel-based finite element analysis." Bone 36(3): 480-8.

Sheyn, D., N. Kimelman-Bleich, et al. (2008). "Ultrasound-based nonviral gene delivery induces bone formation in vivo." Gene Ther 15(4): 257-66.

Shigley, J. E. and C. R. Mischke (2001). Mechanical Engineering Design. New York, McGraw-Hill.

Simonds, R. J., S. D. Holmberg, et al. (1992). "Transmission of human immunodeficiency virus type 1 from a seronegative organ and tissue donor." N Engl J Med 326(11): 726-32.

Soltan, M., D. Smiler, et al. (2007). "Bone block allograft impregnated with bone marrow aspirate." Implant Dent 16(4): 329-39.

Stevenson, S. (1999). "Biology of bone grafts." Orthop Clin North Am 30(4): 543-52. Stevenson, S. and M. Horowitz (1992). "The response to bone allografts." J Bone

Joint Surg Am 74(6): 939-50. Stevenson, S., X. Q. Li, et al. (1997). "Critical biological determinants of

incorporation of non-vascularized cortical bone grafts. Quantification of a complex process and structure." J Bone Joint Surg Am 79(1): 1-16.

Summers, B. N. and S. M. Eisenstein (1989). "Donor site pain from the ilium. A complication of lumbar spine fusion." J Bone Joint Surg Br 71(4): 677-80.

Taddei, F., M. Viceconti, et al. (2003). "Mechanical strength of a femoral reconstruction in paediatric oncology: a finite element study." Proc Inst Mech Eng [H] 217(2): 111-9.

Tam, C. S., J. N. Heersche, et al. (1982). "Parathyroid hormone stimulates the bone apposition rate independently of its resorptive action: differential effects of intermittent and continuous administration." Endocrinology 110(2): 506-12.

Tamhane, A. D., D (2000). Statistics and Data Analysis: from Elementary to Intermediate. Upper Saddle River, Prentice-Hall, Inc.

155

Tan, K. K., G. H. Tan, et al. (2005). "Bone graft substitute using hydroxyapatite scaffold seeded with tissue engineered autologous osteoprogenitor cells in spinal fusion: early result in a sheep model." Med J Malaysia 60 Suppl C: 53-8.

Taschereau, R., P. L. Chow, et al. (2006). "Monte carlo simulations of dose from microCT imaging procedures in a realistic mouse phantom." Med Phys 33(1): 216-24.

Thiele, O. C., C. Eckhardt, et al. (2007). "Factors affecting the stability of screws in human cortical osteoporotic bone: a cadaver study." J Bone Joint Surg Br 89(5): 701-5.

Thompson, R. C., Jr., A. Garg, et al. (2000). "Fractures in large-segment allografts." Clin Orthop Relat Res(370): 227-35.

Thompson, R. C., Jr., E. A. Pickvance, et al. (1993). "Fractures in large-segment allografts." J Bone Joint Surg Am 75(11): 1663-73.

Thurner, P. J., P. Wyss, et al. (2006). "Time-lapsed investigation of three-dimensional failure and damage accumulation in trabecular bone using synchrotron light." Bone 39(2): 289-99.

Tingart, M. J., M. Apreleva, et al. (2003). "The cortical thickness of the proximal humeral diaphysis predicts bone mineral density of the proximal humerus." J Bone Joint Surg Br 85(4): 611-7.

Tiyapatanaputi, P., P. T. Rubery, et al. (2004). "A novel murine segmental femoral graft model." J Orthop Res 22(6): 1254-60.

Tolstunov, L. (2007). "Implant zones of the jaws: implant location and related success rate." J Oral Implantol 33(4): 211-20.

Tsuchida, H., J. Hashimoto, et al. (2003). "Engineered allogeneic mesenchymal stem cells repair femoral segmental defect in rats." J Orthop Res 21(1): 44-53.

Tsuda, H., T. Wada, et al. (2003). "Efficient BMP2 gene transfer and bone formation of mesenchymal stem cells by a fiber-mutant adenoviral vector." Mol Ther 7(3): 354-65.

Urist, M. R. (1965). "Bone: formation by autoinduction." Science 150(698): 893-9. Uzawa, T., M. Hori, et al. (1995). "Comparison of the effects of intermittent and

continuous administration of human parathyroid hormone(1-34) on rat bone." Bone 16(4): 477-84.

Vahle, J. L., M. Sato, et al. (2002). "Skeletal changes in rats given daily subcutaneous injections of recombinant human parathyroid hormone (1-34) for 2 years and relevance to human safety." Toxicol Pathol 30(3): 312-21.

Vahle, J. L., U. Zuehlke, et al. (2008). "Lack of Bone Neoplasms and Persistence of Bone Efficacy in Cynomolgus Macaques Following Long-term Treatment with Teriparatide [rhPTH(1-34)]*." J Bone Miner Res.

Verborgt, O., G. J. Gibson, et al. (2000). "Loss of osteocyte integrity in association with microdamage and bone remodeling after fatigue in vivo." J Bone Miner Res 15(1): 60-7.

156

Verborgt, O., N. A. Tatton, et al. (2002). "Spatial distribution of Bax and Bcl-2 in osteocytes after bone fatigue: complementary roles in bone remodeling regulation?" J Bone Miner Res 17(5): 907-14.

Wang, X., D. B. Masse, et al. (2007). "Detection of trabecular bone microdamage by micro-computed tomography." J Biomech 40(15): 3397-403.

Ward, W. G., M. D. Gautreaux, et al. (2008). "HLA sensitization and allograft bone graft incorporation." Clin Orthop Relat Res 466(8): 1837-48.

Weiland, A. J., T. W. Phillips, et al. (1984). "Bone grafts: a radiologic, histologic, and biomechanical model comparing autografts, allografts, and free vascularized bone grafts." Plast Reconstr Surg 74(3): 368-79.

Wheeler, D. L. and W. F. Enneking (2005). "Allograft bone decreases in strength in vivo over time." Clin Orthop Relat Res(435): 36-42.

Wheeler, D. L., J. L. Haynie, et al. (2001). "Biomechanical evaluation of retrieved massive allografts: preliminary results." Biomed Sci Instrum 37: 251-6.

Whitfield, J. F. (2005). "Parathyroid hormone (PTH) and hematopoiesis: new support for some old observations." J Cell Biochem 96(2): 278-84.

Wunder, C. C. and R. C. Welch (1977). "Femur-bending properties as influenced by gravity: II. Ultimate load, moment, and stress for 3-G mice." Aviat Space Environ Med 48(8): 734-6.

Xie, C., D. Reynolds, et al. (2007). "Structural bone allograft combined with genetically engineered mesenchymal stem cells as a novel platform for bone tissue engineering." Tissue Eng 13(3): 435-45.

Yazici, C., L. Yanoso, et al. (2008). "The effect of surface demineralization of cortical bone allograft on the properties of recombinant adeno-associated virus coatings." Biomaterials 29(28): 3882-7.

Younger, E. M. and M. W. Chapman (1989). "Morbidity at bone graft donor sites." J Orthop Trauma 3(3): 192-5.

Zhang, X., A. Naik, et al. (2005). "Periosteal stem cells are essential for bone revitalization and repair." J Musculoskelet Neuronal Interact 5(4): 360-2.

Zhang, X., C. Xie, et al. (2005). "Periosteal progenitor cell fate in segmental cortical bone graft transplantations: implications for functional tissue engineering." J Bone Miner Res 20(12): 2124-37.

Zhou, H., A. Iida-Klein, et al. (2003). "Anabolic action of parathyroid hormone on cortical and cancellous bone differs between axial and appendicular skeletal sites in mice." Bone 32(5): 513-20.

Zioupos, P., J. D. Currey, et al. (1995). "Experimentally determined microcracking around a circular hole in a flat plate of bone: comparison with predicted stresses." Philos Trans R Soc Lond B Biol Sci 347(1322): 383-96.

157

7 Appendicies 7.1 APPENDIX A: UNIONRATIO & UCTANALYSIS MATLAB CODE

The critical components of the UnionRatio analysis from the uCTanalysis MATLAB program are presented in this appendix. Below is a screen shot from the program in its entirety.

158

Snake Morphing – Edge Detection. The "Snake This Slice" button function Snake2D(hObject, eventdata, handles) im = handles.currentdata; image_num = handles.imnum; %the 3D image dataset if ~(handles.LastModifiedContours(end) == image_num) handles.LastModifiedContours(end+1) = image_num; end guidata(hObject,handles); mingray = handles.mingraya; maxgray = handles.maxgrayb; M(3) = handles.max; M(2) = handles.x; M(1) = handles.y; im2 = imfilter(im(:,:,image_num),fspecial('gaussian',5,1.8)); % SYNTAX: GradientSum = zeros(M(1),M(2)); GradNeighbors = 3; % this allows the gradient from the neighboring slices to be considered, reducing out of plane noise for i = 0:GradNeighbors * 2 im_Temp = imfilter(im(:,:,image_num-GradNeighbors+i),fspecial('gaussian',5,1.8)); % SYNTAX: fspecial('gaussian', hsize, StandardDeviation); imPrewittVert = filter2(fspecial('prewitt'),im_Temp); % im(:,:,image_num)); imPrewittHoriz = filter2(fspecial('prewitt')',im_Temp); % im(:,:,image_num)); imPrewittCombinedTemp = abs(imPrewittVert) + abs(imPrewittHoriz); GradientSum = GradientSum + imPrewittCombinedTemp; end imPrewittCombined = GradientSum; xOUT = handles.SliceContours{image_num,1}(:,1); %xC1OUT yOUT = handles.SliceContours{image_num,1}(:,2); %yC1OUT [xOUT, yOUT] = SubroutineA(hObject, eventdata, handles, xOUT, yOUT, im2, mingray, imPrewittCombined); handles.SliceContours{image_num,1}(:,1) = xOUT; handles.SliceContours{image_num,1}(:,2) = yOUT; guidata(hObject,handles); [xSOUT,ySOUT]=splineDGR(xOUT,yOUT); if ~isempty(handles.SliceContours{image_num,2}); xINN = handles.SliceContours{image_num,2}(:,1); yINN = handles.SliceContours{image_num,2}(:,2); [xINN, yINN] = SubroutineA(hObject, eventdata, handles, xINN, yINN, im2, mingray, imPrewittCombined); handles.SliceContours{image_num,2}(:,1) = xINN; handles.SliceContours{image_num,2}(:,2) = yINN; guidata(hObject,handles); [xSINN,ySINN]=splineDGR(xINN,yINN); end axes(handles.CT); imagesc(im(:,:,image_num),[(mingray) (maxgray)]) axis equal hold on plot([xSOUT;xSOUT(1)],[ySOUT;ySOUT(1)],'g-',[xOUT],[yOUT],'go'); if ~isempty(handles.SliceContours{image_num,2}); plot([xSINN;xSINN(1)],[ySINN;ySINN(1)],'r-',[xINN],[yINN],'ro'); end hold off handles.UpdatedContoursWithoutSave = 1; guidata(hObject, handles) DilateSnake2D(hObject, eventdata, handles); % % % % % % % % % % THE GUTS OF THE PROGRAM % % % % % % % % % % % % function [xs, ys] = SubroutineA(hObject, eventdata, handles, xs, ys, im2, mingray, imPrewittCombined) % find the slope of a point by looking at previous and next points slopes(1) = (ys(2) - ys(length(ys))) / (xs(2) - xs(length(xs))); % for first point slopes(length(xs)) = (ys(length(ys)-1) - ys(1)) / (xs(length(xs)-1) - xs(1)); % for last point for i = 2:(length(xs)-1) slopes(i) = (ys(i+1) - ys(i-1)) / (xs(i+1) - xs(i-1)); end % use negative reciprocal to find the slope of the perpendicular at a point perp = -1./slopes; % find the unit-length in the X and Y directions for a line ix = sign(perp)*1./(sqrt(1+perp.^2)); iy = sqrt(1-ix.^2); PerpLength = str2num(get(handles.EdgeSearchDistEditTextbox, 'String')); for j = 1:length(xs) for i = -PerpLength:PerpLength PerpLinesX(j,(i+PerpLength+1)) = xs(j)+i*ix(j); PerpLinesY(j,(i+PerpLength+1)) = ys(j)+i*iy(j); end

159

end % Find Maximum Grayscale Intensity from Gradient Image PerpLinesX; PerpLinesY; for j = 1:length(xs) % j is the point on the contour being evaluated individually for i = 1:(PerpLength*2+1) PerpLineGradGrayscales(j,i) = imPrewittCombined(round(PerpLinesY(j,i)),round(PerpLinesX(j,i))); % imPrewittCombinedMat end for i = 1:(PerpLength*2+1) PerpLineImGrayscales(j,i) = im2(round(PerpLinesY(j,i)),round(PerpLinesX(j,i))); end PerpLineGradGrayscales(j,:); MaxGradGrayscale = max(PerpLineGradGrayscales(j,:)); IndexMaxGradGrayscale = find(MaxGradGrayscale == PerpLineGradGrayscales(j,:)); xs_ImageE(j) = PerpLinesX(j,IndexMaxGradGrayscale(1)); ys_ImageE(j) = PerpLinesY(j,IndexMaxGradGrayscale(1)); % plot(xs(j),ys(j), 'b*'); % Now Measure the change in image energy Del_Image_E = abs(MaxGradGrayscale - PerpLineGradGrayscales(j,(PerpLength+1))); % (PerpLength+1) is the original digitized position % Now Measure the change in spring energy K = 1000; % Spring Constant if j == 1 OrigDistanceLeft = sqrt( (xs(j)-xs(length(xs)))^2 + (ys(j)-ys(length(xs)))^2 ); NewDistanceLeft = sqrt( (xs_ImageE(j)-xs(length(xs)))^2 + (ys_ImageE(j)-ys(length(xs)))^2 ); Del_DistanceLeft = OrigDistanceLeft - NewDistanceLeft; OrigDistanceRight = sqrt( (xs(j)-xs(j+1))^2 + (ys(j)-ys(j+1))^2 ); NewDistanceRight = sqrt( (xs_ImageE(j)-xs(j+1))^2 + (ys_ImageE(j)-ys(j+1))^2 ); elseif j == (length(xs)) OrigDistanceLeft = sqrt( (xs(j)-xs(j-1))^2 + (ys(j)-ys(j-1))^2 ); NewDistanceLeft = sqrt( (xs_ImageE(j)-xs(j-1))^2 + (ys_ImageE(j)-ys(j-1))^2 ); Del_DistanceLeft = OrigDistanceLeft - NewDistanceLeft; OrigDistanceRight = sqrt( (xs(j)-xs(1))^2 + (ys(j)-ys(1))^2 ); NewDistanceRight = sqrt( (xs_ImageE(j)-xs(1))^2 + (ys_ImageE(j)-ys_ImageE(1))^2 ); else OrigDistanceLeft = sqrt( (xs(j)-xs(j-1))^2 + (ys(j)-ys(j-1))^2 ); NewDistanceLeft = sqrt( (xs_ImageE(j)-xs(j-1))^2 + (ys_ImageE(j)-ys(j-1))^2 ); Del_DistanceLeft = OrigDistanceLeft - NewDistanceLeft; OrigDistanceRight = sqrt( (xs(j)-xs(j+1))^2 + (ys(j)-ys(j+1))^2 ); NewDistanceRight = sqrt( (xs_ImageE(j)-xs(j+1))^2 + (ys_ImageE(j)-ys(j+1))^2 ); end Del_DistanceRight = OrigDistanceRight - NewDistanceRight; Del_Spring_E = abs(K*( Del_DistanceLeft + Del_DistanceRight ) ); if (Del_Image_E > Del_Spring_E) % Now, Find out if any of the Perp line grayscales fall within Bone threshold % If they don't, don't bother moving the point if max(PerpLineImGrayscales(j,:)) < mingray xs_ImageE(j) = xs(j); end xs(j) = xs_ImageE(j); ys(j) = ys_ImageE(j); end end

Snake Morphing – Dilation: function DilateSnake2D(hObject, eventdata, handles) im = handles.currentdata; image_num = handles.imnum; mingray = handles.mingraya; maxgray = handles.maxgrayb; M(3) = handles.max; M(2) = handles.x; M(1) = handles.y; im2 = imfilter(im(:,:,image_num),fspecial('gaussian',5,1.8)); % SYNTAX: fspecial('gaussian', hsize, StandardDeviation); xOUT = handles.SliceContours{image_num,1}(:,1); %xC1OUT yOUT = handles.SliceContours{image_num,1}(:,2); %yC1OUT

160

[xOUT,yOUT]=SubroutineA(hObject, eventdata, handles, xOUT, yOUT, im2, mingray); handles.SliceContours{image_num,1}(:,1) = xOUT; handles.SliceContours{image_num,1}(:,2) = yOUT; guidata(hObject,handles); [xSOUT,ySOUT]=splineDGR(xOUT,yOUT); if ~isempty(handles.SliceContours{image_num,2}); xINN = handles.SliceContours{image_num,2}(:,1); yINN = handles.SliceContours{image_num,2}(:,2); [xINN,yINN]=SubroutineA(hObject, eventdata, handles, xINN, yINN, im2, mingray); handles.SliceContours{image_num,2}(:,1) = xINN; handles.SliceContours{image_num,2}(:,2) = yINN; guidata(hObject,handles); [xSINN,ySINN]=splineDGR(xINN,yINN); end % plot(xs,ys, 'b*'); % hold off axes(handles.CT); imagesc(im(:,:,image_num),[(mingray) (maxgray)]) axis equal hold on plot([xSOUT;xSOUT(1)],[ySOUT;ySOUT(1)],'g-',[xOUT],[yOUT],'go'); if ~isempty(handles.SliceContours{image_num,2}); plot([xSINN;xSINN(1)],[ySINN;ySINN(1)],'r-',[xINN],[yINN],'ro'); end hold off % % % % % % % % % % THE GUTS OF THE PROGRAM % % % % % % % % % % % % function [xs ,ys ] = SubroutineA(hObject, eventdata, handles, xs, ys, im2, mingray) slopes(1) = (ys(2) - ys(length(ys))) / (xs(2) - xs(length(xs))); % for first point slopes(length(xs)) = (ys(length(ys)-1) - ys(1)) / (xs(length(xs)-1) - xs(1)); % for last point for i = 2:(length(xs)-1) slopes(i) = (ys(i+1) - ys(i-1)) / (xs(i+1) - xs(i-1)); end % use negative reciprocal to find the slope of the perpendicular at a point perp = -1./slopes; % find the unit-length in the X and Y directions for a line ix = sign(perp)*1./(sqrt(1+perp.^2)); iy = sqrt(1-ix.^2); PerpLength = str2num(get(handles.DilateSearchDistEditTextbox,'String')); for j = 1:length(xs) for i = -PerpLength:PerpLength PerpLinesX(j,(i+PerpLength+1)) = xs(j)+i*ix(j); PerpLinesY(j,(i+PerpLength+1)) = ys(j)+i*iy(j); end end % hold on % plot(PerpLinesX', PerpLinesY', 'r-'); % plot perpendicular lines at each point in red % Find Minimum Grayscale Intensity from ORIGINAL Image for j = 1:length(xs) for i = 1:(PerpLength*2+1) PerpLineGrayscales(j,i) = im2(round(PerpLinesY(j,i)),round(PerpLinesX(j,i))); end for i = 1:(PerpLength*2+1) PerpLineImGrayscales(j,i) = im2(round(PerpLinesY(j,i)),round(PerpLinesX(j,i))); end PerpLineGrayscales(j,:); MinGrayscale = min(PerpLineGrayscales(j,:)); IndexMinGrayscale = find(MinGrayscale == PerpLineGrayscales(j,:)); xs_ImageE(j) = PerpLinesX(j,IndexMinGrayscale(1)); ys_ImageE(j) = PerpLinesY(j,IndexMinGrayscale(1)); % plot(xs(j),ys(j), 'b*'); if max(PerpLineImGrayscales(j,:)) < mingray xs_ImageE(j) = xs(j); end xs(j) = xs_ImageE(j); ys(j) = ys_ImageE(j); end

Surface calculation of UnionRatio. "Connectivity Surface Area" button

161

function findsurfaceareaDave(hObject,handles) %The function findsurfacearea takes the contours of all the slices which %appear in the listbox and finds the surface area of the 3-D surface that %the contours create when they are passed through the Matlab function %isosurface. This is accomplsihed by using the faces and vertices output of %isosurface. %Findsurfacearea also finds the surface area of the end caps of the 3-D %surface created by the passing the entire contour data set through the %function isosurface. This is accomplished by using the function isocaps %and its face and vertices output. %Findsurface area also finds the surface area of connection of a contoured %object to another object. This accomplished by summing 1/3 of the area of %one face of the connected surface if that face is in contact with a pixel %that has a grayscale value above a specific threshold value. %The surface area of the contoured object and the surface area of %connection area converted to milimeters squared and outputted to the %command window. %The function findsurfacearea seperates the contoured regions into a top %region, a middle region and a bottom region. This is done to conserve %memory usage. % load all necessary data from handles structure im = handles.currentdata; image_num = handles.imnum; mingray = handles.mingraya; maxgray = handles.maxgrayb; mingray = {num2str(mingray)}; Mz = handles.max; Mx = handles.x; My = handles.y; Ratio = handles.Resolution3D(3)/handles.Resolution3D(1); pad = 5; %eliminate zeros from slice data getslices = cell2struct(get(handles.ContouredSlicesListbox,'String'),'Slices',2); current_im_slice = str2num(getslices(1).Slices); true_first_slice = str2num(getslices(1).Slices); last_slice = str2num(getslices(end).Slices); %extract corresponding contour data to slice data contours = handles.SliceContours(true_first_slice:last_slice,:); slices_length = length(contours); %seperate out x and y coordinates of contour data for i = 1:(last_slice+1-true_first_slice) xdataOUT{i} = contours{i,1}(:,1); ydataOUT{i} = contours{i,1}(:,2); if ~isempty(contours{i,2}); xdataINN{i} = contours{i,2}(:,1); ydataINN{i} = contours{i,2}(:,2); end end %find the smallest increment that the length of contour array is divisible %by, so that the stack of contours may be broken down into top, middle and %bottom region. This allows for smaller volumes to be created in each %region (especially the middle region)in order to conserve memory during %the actual surface area calculation SliceDivisNum = 17; while (mod((last_slice-true_first_slice),SliceDivisNum) ~= 0) % mod = Modulus after division. (remainder) SliceDivisNum = SliceDivisNum - 1; end if SliceDivisNum == 1 msgbox('Number of slices is a prime number') return else SliceDivisNum; end mingray = round(str2num(get(handles.BinaryEditText, 'String'))); TextForInputDlg = ['SliceDivisNum is ', num2str(SliceDivisNum), '. EnterThresholdValue.' ]; mingray = inputdlg(TextForInputDlg,'threshold',1,{num2str(mingray)}); % if 'cancel' is clicked if isempty(mingray) return else mingray = str2num(cell2mat(mingray)); end set(handles.MinGraySelector, 'Value', mingray); set(handles.MinGrayText, 'String',['Min Gray: ' num2str(mingray)]); handles.mingraya = mingray; guidata(hObject,handles);

162

contourarea_final = double(0); connectedarea_final = double(0); areamineral_final = double(0); contourarea_array = zeros(length(contours),1); connectedarea_array = zeros(length(contours),1); areamineral_array = zeros(length(contours),1); h = waitbar(0,'Calculating Connected Surface Area. Please wait...'); start = 1; %start is in the binary mask's reference frame (BW) while first_slice is the global reference frame (im). finish = start+pad; %start and finish are used to iterate through the contour data array i = 0; %counter to used to create a 3-D binary mask for a the regions % make the binary mask for all the slices getsliceLength = length(getslices); TotalBW = zeros(My,Mx,getsliceLength+2*pad); % Pad the top of the binary mask with Zeros i=pad; for j = 1:(getsliceLength) i = i + 1; % pad + 1 slice = str2num(getslices(j).Slices); xOUT = cell2mat(xdataOUT(j)); yOUT = cell2mat(ydataOUT(j)); [x2OUT,y2OUT] = splineDGR(xOUT,yOUT); BWOUT = roipolyoldDave(im(:,:,slice),x2OUT,y2OUT); if ~isempty(handles.SliceContours{true_first_slice+j-1,2}); xINN = handles.SliceContours{true_first_slice+j-1,2}(:,1); yINN = handles.SliceContours{true_first_slice+j-1,2}(:,2); [x2INN,y2INN] = splineDGR(xINN,yINN); BWINN = roipolyoldDave(im(:,:,slice),x2INN,y2INN); BWOUT = BWINN ~= BWOUT; end TotalBW(:,:,i) = BWOUT; % figure; imagesc(BWtop(:,:,i)); end TotalBWsize = size(TotalBW); %TotalBW(:,:,TotalBWsize(3):(TotalBWsize(3)+pad)) = zeros(Mx,My,pad); % Pad the bottom of the binary mask with Zeros % Pad the top of the binary mask with Zeros to measure the % connection at the top surface (Dave) current_im_slice = current_im_slice-pad; % TotalBW = cat(3,zeros(TotalBWsize(1),TotalBWsize(2),pad),TotalBW); % Pad the bottom of the binary mask with Zeros % TotalBW = cat(3,TotalBW,zeros(TotalBWsize(1),TotalBWsize(2),pad)); %code for smoothing BW using a guassian filter TotalBW = smooth3(TotalBW,'gaussian',[pad pad (round(0.5*pad/Ratio)+0.5)*2], 2.0); % % TotalBW = smooth3(TotalBW, 'box', [3,3,3]); % % TotalBW = smooth3(TotalBW,'gaussian',[pad pad 3], 2.0); % TotalBW = TotalBW(:,:,:); %% Perform Top Cap measurement outside the loop. % Top Cap will now be the length of the pad that was applied: BWtop = TotalBW(:,:,1:pad+1); waitbar(pad/(slices_length+pad*2),h); BWtopsize = size(BWtop); x = 0; y = 0; z = 0; [x,y,z] = meshgrid(1:BWtopsize(2),1:BWtopsize(1),1:BWtopsize(3)); fv = isosurface(x,y,z,BWtop,0.5); %from the image processing toolbox fv.CDataFaces = ones(length(fv.faces),1); for i = 1:length(fv.faces) v1 = fv.vertices(fv.faces(i,1),1); v2 = fv.vertices(fv.faces(i,1),2); v3 = fv.vertices(fv.faces(i,1),3); if im(round(v2),round(v1),(round(v3+current_im_slice-1))) >= mingray fv.CDataFaces(i) = im(round(v2),round(v1),(round(v3+current_im_slice-1))); end end fv.CDataVertices = ones(length(fv.vertices),1); for i = 1:length(fv.vertices) v1 = fv.vertices(i,1); v2 = fv.vertices(i,2); v3 = fv.vertices(i,3); if im(round(v2),round(v1),(round(v3+current_im_slice-1))) >= mingray fv.CDataVertices(i) = im(round(v2),round(v1),(round(v3+current_im_slice-1))); % 255

163

% fv.CDataVertices(i) = 255; end end % This moves the verticies in the z-direction from the local % reference plane to the global: TempVertices = fv.vertices; c = zeros(length(TempVertices),2); c = cat(2,c,(ones(length(TempVertices),1)*(current_im_slice))); % -1 TempVertices = TempVertices + c; fv_total.vertices = TempVertices; fv_total.faces = fv.faces; fv_total.CDataFaces = fv.CDataFaces; fv_total.CDataVertices = fv.CDataVertices; %pull out faces and vertices from isosurface vertexpointstop = fv.vertices; facetop = fv.faces; Contour_Area = double(0); Connected_Area = double(0); AreaMineral = double(0); A = 0; %area of shell (from isosurface) for m = 1:length(facetop(:,1)) vertices = facetop(m,:); v1 = vertexpointstop(vertices(1),:); v2 = vertexpointstop(vertices(2),:); v3 = vertexpointstop(vertices(3),:); v1a = v1; v2a = v3; v3a = v2; v1a(3) = v1(3)*Ratio; v2a(3) = v3(3)*Ratio; v3a(3) = v2(3)*Ratio; %base v1 and v2 base = sqrt(((v2a(1)-v1a(1)).^2)+((v2a(2)-v1a(2)).^2)+((v2a(3)-v1a(3)).^2)); %vector between v1 and v2 vect1 = [v2a(1)-v1a(1),v2a(2)-v1a(2),v2a(3)-v1a(3)]; %vector between v1 and v2 vect2 = [v3a(1)-v1a(1),v3a(2)-v1a(2),v3a(3)-v1a(3)]; %distance to point 3 c = cross(vect1,vect2); d = sqrt(dot(c,c))/sqrt(dot(vect1,vect1)); A = (0.5*base*d); Contour_Area = Contour_Area + A; %connected area of shell (from isosurface) if im(round(v1(2)),round(v1(1)),(round(v1(3)+current_im_slice-1))) >= mingray Connected_Area = Connected_Area + (1/3*A); AreaMineral = AreaMineral + ... (1/3*A)*double(im(round(v1(2)),round(v1(1)),(round(v1(3)+current_im_slice-1)))*0.10197 - 232); end if im(round(v2(2)),round(v2(1)),(round(v2(3)+current_im_slice-1))) >= mingray Connected_Area = Connected_Area + (1/3*A); AreaMineral = AreaMineral + ... (1/3*A)*double(im(round(v1(2)),round(v1(1)),(round(v1(3)+current_im_slice-1)))*0.10197 - 232); end if im(round(v3(2)),round(v3(1)),(round(v3(3)+current_im_slice-1))) >= mingray Connected_Area = Connected_Area + (1/3*A); AreaMineral = AreaMineral + ... (1/3*A)*double(im(round(v1(2)),round(v1(1)),(round(v1(3)+current_im_slice-1)))*0.10197 - 232); end end %sum contoured and connected area of top region into final %contoured and connected area of entire object contourarea_final = contourarea_final + Contour_Area; contourarea_array(start) = Contour_Area; connectedarea_array(start) = Connected_Area; connectedarea_final = connectedarea_final + Connected_Area;

164

areamineral_array(start) = AreaMineral; areamineral_final = areamineral_final + AreaMineral; contourarea_TopCap = Contour_Area; connectedarea_TopCap = Connected_Area; areamineral_TopCap = AreaMineral; %iterate through slices by increment 'SliceDivisNum' into next region current_im_slice = current_im_slice+pad; %increment start and finish by the increment 'SliceDivisNum' found outside %the while loop so that can proceed into the next region (middle %region) start = pad+1; finish = start+SliceDivisNum; %% Perform Middle Region surface area calculation inside while loop while current_im_slice <= (last_slice+pad) if (current_im_slice < last_slice) waitbar(start/(slices_length+pad*2),h) BW = TotalBW(:,:,start:finish); BWsize = size(BW); x = 0; y = 0; z = 0; [x,y,z] = meshgrid(1:BWsize(2),1:BWsize(1),1:BWsize(3)); fv = isosurface(x,y,z,BW,0.5); % fv = reducepatch(fv,0.5); fv.CDataFaces = ones(length(fv.faces),1); for i = 1:length(fv.faces) % fv.faces(i,1), fv.faces(i,2), fv.faces(i,3) v1 = fv.vertices(fv.faces(i,1),1); v2 = fv.vertices(fv.faces(i,1),2); v3 = fv.vertices(fv.faces(i,1),3); if im(round(v2),round(v1),(round(v3+current_im_slice-1))) >= mingray fv.CDataFaces(i) = im(round(v2),round(v1),(round(v3+current_im_slice-1))); % 255 end end fv.CDataVertices = ones(length(fv.vertices),1); for i = 1:length(fv.vertices) % fv.faces(i,1), fv.faces(i,2), fv.faces(i,3) v1 = fv.vertices(i,1); v2 = fv.vertices(i,2); v3 = fv.vertices(i,3); if im(round(v2),round(v1),(round(v3+current_im_slice-1))) >= mingray fv.CDataVertices(i) = im(round(v2),round(v1),(round(v3+current_im_slice-1))); % 255 % fv.CDataVertices(i) = 255; end end NumberOfVertices = length(fv_total.vertices); % Code to modify the fv vertices' Z-coordinate such that it matches the Image data Z-coordinate: TempVertices = fv.vertices; c = zeros(length(TempVertices),2); c = cat(2,c,(ones(length(TempVertices),1)*current_im_slice)); TempVertices = TempVertices + c; fv_total.vertices = cat(1, fv_total.vertices, TempVertices); fv_total.faces = cat(1, fv_total.faces, (fv.faces + NumberOfVertices)); fv_total.CDataFaces = cat(1, fv_total.CDataFaces, fv.CDataFaces); fv_total.CDataVertices = cat(1, fv_total.CDataVertices, fv.CDataVertices); vertexpoints = fv.vertices; face = fv.faces; Contour_Area = double(0); Connected_Area = double(0); AreaMineral = double(0); A = 0; %area of shell (from isosurface) for m = 1:length(face(:,1)) vertices = face(m,:); v1 = vertexpoints(vertices(1),:); v2 = vertexpoints(vertices(2),:); v3 = vertexpoints(vertices(3),:);

165

v1a = v1; v2a = v3; v3a = v2; v1a(3) = v1(3)*Ratio; v2a(3) = v3(3)*Ratio; v3a(3) = v2(3)*Ratio; %base v1 and v2 base = sqrt(((v2a(1)-v1a(1)).^2)+((v2a(2)-v1a(2)).^2)+((v2a(3)-v1a(3)).^2)); %vector between v1 and v2 vect1 = [v2a(1)-v1a(1),v2a(2)-v1a(2),v2a(3)-v1a(3)]; %vector between v1 and v2 vect2 = [v3a(1)-v1a(1),v3a(2)-v1a(2),v3a(3)-v1a(3)]; %distance to point 3 c = cross(vect1,vect2); d = sqrt(dot(c,c))/sqrt(dot(vect1,vect1)); A = (0.5*base*d); Contour_Area = Contour_Area + A; %connected area of middle region (from isosurface) if im(round(v1(2)),round(v1(1)),(round(v1(3)+current_im_slice-1))) >= mingray Connected_Area = Connected_Area + (1/3*A); AreaMineral = AreaMineral + ... (1/3*A)*double(im(round(v1(2)),round(v1(1)),(round(v1(3)+current_im_slice-1)))*0.10197 - 232); end if im(round(v2(2)),round(v2(1)),(round(v2(3)+current_im_slice-1))) >= mingray Connected_Area = Connected_Area + (1/3*A); AreaMineral = AreaMineral + ... (1/3*A)*double(im(round(v1(2)),round(v1(1)),(round(v1(3)+current_im_slice-1)))*0.10197 - 232); end if im(round(v3(2)),round(v3(1)),(round(v3(3)+current_im_slice-1))) >= mingray Connected_Area = Connected_Area + (1/3*A); AreaMineral = AreaMineral + ... (1/3*A)*double(im(round(v1(2)),round(v1(1)),(round(v1(3)+current_im_slice-1)))*0.10197 - 232); end end clear('vertexpoints'); clear('face'); clear('TempVertices'); contourarea_final = contourarea_final + Contour_Area; connectedarea_final = connectedarea_final + Connected_Area; areamineral_final = areamineral_final + AreaMineral; contourarea_array(start) = Contour_Area; connectedarea_array(start) = Connected_Area; areamineral_array(start) = AreaMineral; current_im_slice = current_im_slice + SliceDivisNum; start = finish; finish = start+SliceDivisNum; else %% %%%%%%%%% Bottom Region with Bottom Cap %%%%%%%%%%% % make sure that finish does not exceed length of slice stack and % contour stack finish = length(TotalBW); i = 0; waitbar(start/(slices_length+pad*2),h) BWbot = TotalBW(:,:,start:end); BWbotsize = size(BWbot); x = 0; y = 0; z = 0; [x,y,z] = meshgrid(1:BWbotsize(2),1:BWbotsize(1),1:BWbotsize(3)); fv = isosurface(x,y,z,BWbot,0.5); % fv = reducepatch(fv,0.5); fv.CDataFaces = ones(length(fv.faces),1); for i = 1:length(fv.faces) % fv.faces(i,1), fv.faces(i,2), fv.faces(i,3) v1 = fv.vertices(fv.faces(i,1),1); v2 = fv.vertices(fv.faces(i,1),2); v3 = fv.vertices(fv.faces(i,1),3); if im(round(v2),round(v1),(round(v3+current_im_slice-1))) >= mingray fv.CDataFaces(i) = im(round(v2),round(v1),(round(v3+current_im_slice-1))); % 255 end end

166

fv.CDataVertices = ones(length(fv.vertices),1); for i = 1:length(fv.vertices) % fv.faces(i,1), fv.faces(i,2), fv.faces(i,3) v1 = fv.vertices(i,1); v2 = fv.vertices(i,2); v3 = fv.vertices(i,3); if im(round(v2),round(v1),(round(v3+current_im_slice-1))) >= mingray fv.CDataVertices(i) = im(round(v2),round(v1),(round(v3+current_im_slice-1))); % 255 % fv.CDataVertices(i) = 255; end end % Code to modify the fv vertices' Z-coordinate such that it matches the Image data Z-coordinate: TempVertices = fv.vertices; c = zeros(length(TempVertices),2); c = cat(2,c,(ones(length(TempVertices),1)*current_im_slice)); TempVertices = TempVertices + c; NumberOfVertices = length(fv_total.vertices); fv_total.vertices = cat(1, fv_total.vertices, TempVertices); fv_total.faces = cat(1, fv_total.faces, (fv.faces + NumberOfVertices)); fv_total.CDataFaces = cat(1, fv_total.CDataFaces, fv.CDataFaces); fv_total.CDataVertices = cat(1, fv_total.CDataVertices, fv.CDataVertices); % BoneSurfaceFV = handles.BoneSurfaceFV; % BoneSurfaceFV.vertices(:,3) = BoneSurfaceFV.vertices(:,3) * Ratio; fv_total.vertices(:,3) = (fv_total.vertices(:,3) * Ratio-1); handles.UnionAreaSurfaceFV = fv_total; guidata(hObject, handles); figure; % fv_total.CDataVertices = fv_total.CDataVertices + min(fv_total.CDataVertices); p_fv = patch('Faces',fv_total.faces,'Vertices',fv_total.vertices,'FaceVertexCData',fv_total.CDataVertices, 'FaceColor','interp', 'edgecolor','none'); % 'FaceColor', 'flat' axis equal, axis tight, lighting phong, % view(215,-25), camlight, view(315,25), % camlight c = colormap('jet'); rotate3d % colormap gray; % clength = length(c); % c(1:clength/2,1)=.0; c(1:clength/2,2)=.0; c(1:clength/2,3)=.7; c(clength/2+1:clength,1)=.7; c(clength/2+1:clength,2)=.0; c(clength/2+1:clength,3)=.0; % colormap(c); global lighthandle; lighthandle=[]; uicontrol('Style', 'pushbutton', 'String', 'Light',... 'Position', [10 10 50 30], 'Callback', 'UASurfaceLighting()'); vertexpointsbot = fv.vertices; facebot = fv.faces; Contour_Area = double(0); Connected_Area = double(0); AreaMineral = double(0); A = 0; %area of shell(from isosurface) for m = 1:length(facebot(:,1)) vertices = facebot(m,:); v1 = vertexpointsbot(vertices(1),:); v2 = vertexpointsbot(vertices(2),:); v3 = vertexpointsbot(vertices(3),:); v1a = v1; v2a = v3; v3a = v2; v1a(3) = v1(3)*Ratio; v2a(3) = v3(3)*Ratio; v3a(3) = v2(3)*Ratio; %base v1 and v2 base = sqrt(((v2a(1)-v1a(1)).^2)+((v2a(2)-v1a(2)).^2)+((v2a(3)-v1a(3)).^2)); %vector between v1 and v2 vect1 = [v2a(1)-v1a(1),v2a(2)-v1a(2),v2a(3)-v1a(3)]; %vector between v1 and v2 vect2 = [v3a(1)-v1a(1),v3a(2)-v1a(2),v3a(3)-v1a(3)]; %distance to point 3

167

c = cross(vect1,vect2); d = sqrt(dot(c,c))/sqrt(dot(vect1,vect1)); A = (0.5*base*d); Contour_Area = Contour_Area + A; %connected area of bottom region (from isosurface) if im(round(v1(2)),round(v1(1)),(round(v1(3)+current_im_slice-1))) >= mingray Connected_Area = Connected_Area + (1/3*A); AreaMineral = AreaMineral + ... (1/3*A)*double(im(round(v1(2)),round(v1(1)),(round(v1(3)+current_im_slice-1)))*0.10197 - 232); end if im(round(v2(2)),round(v2(1)),(round(v2(3)+current_im_slice-1))) >= mingray Connected_Area = Connected_Area + (1/3*A); AreaMineral = AreaMineral + ... (1/3*A)*double(im(round(v1(2)),round(v1(1)),(round(v1(3)+current_im_slice-1)))*0.10197 - 232); end if im(round(v3(2)),round(v3(1)),(round(v3(3)+current_im_slice-1))) >= mingray Connected_Area = Connected_Area + (1/3*A); AreaMineral = AreaMineral + ... (1/3*A)*double(im(round(v1(2)),round(v1(1)),(round(v1(3)+current_im_slice-1)))*0.10197 - 232); end end current_im_slice = current_im_slice + SliceDivisNum; %iterate to end while loop data(1,:) = {'Contour Area No bottom cap (mm2)', 'Connected Area No bottom cap (mm2)','Contour Area No Top cap (mm2)', 'Connected Area No Top cap (mm2)',... 'TopHalfContourArea', 'TopHalfConnectedArea', 'BottomHalfContourArea', 'BottomHalfConnectedArea', 'WholeContourArea','WholeConnectedArea',... 'TopRatio','BottomRatio','MinimumConnectedRatio', 'Threshold', 'TopHalfAreaMineral [mg/cc * mm^2]', 'BottomHalfAreaMineral [mg/cc * mm^2]', 'AreaMineral [mg/cc * mm^2]','MineralDensity of Union [mg/cc]'}; disp('contour Area without bottom cap:') Resolution = handles.Resolution; contourarea_finalmm = contourarea_final*(Resolution^2) connectedarea_finalmm = connectedarea_final*(Resolution^2) data(2,1:2) = {sprintf('%0.4f', contourarea_finalmm),sprintf('%0.4f', connectedarea_finalmm)}; contourarea_final = contourarea_final + Contour_Area; connectedarea_final = connectedarea_final + Connected_Area; areamineral_final = areamineral_final + AreaMineral; contourarea_array(start) = Contour_Area; connectedarea_array(start) = Connected_Area; %adds bottom cap areamineral_array(start) = AreaMineral; end end Resolution = handles.Resolution; disp('contour Area without top cap:') contourarea_NoTopCap = contourarea_final - contourarea_TopCap; connectedarea_NoTopCap = connectedarea_final - connectedarea_TopCap; areamineral_NoTopCap = areamineral_final - areamineral_TopCap; contourarea_NoTopCapmm = contourarea_NoTopCap*(Resolution^2) connectedarea_NoTopCapmm = connectedarea_NoTopCap*(Resolution^2) areamineral_NoTopCapmm = areamineral_NoTopCap*(Resolution^2) disp('contour area WITH both caps:') contourarea_finalmm = contourarea_final*(Resolution^2) connectedarea_finalmm = connectedarea_final*(Resolution^2) areamineral_finalmm = areamineral_final*(Resolution^2) %outputs to the command window of final contour are in milimeters squared %and final connected area in milimeters squared disp('Connectivity when contour is complete, and splitting connectivity in halves:') TopHalfContourArea = sum(contourarea_array(1:round(end/2)))*(Resolution^2) TopHalfConnectedArea = sum(connectedarea_array(1:round(end/2)))*(Resolution^2) BottomHalfContourArea = sum(contourarea_array(round(end/2):end))*(Resolution^2) BottomHalfConnectedArea = sum(connectedarea_array(round(end/2):end))*(Resolution^2) WholeContourArea = sum(contourarea_array(1:end))*(Resolution^2) WholeConnectedArea = sum(connectedarea_array(1:end))*(Resolution^2)

168

WholeAreaMineral = sum(areamineral_array(1:end))*(Resolution^2) TopRatio = TopHalfConnectedArea/TopHalfContourArea BottomRatio = BottomHalfConnectedArea/BottomHalfContourArea MinimumConnectedRatio = min(TopRatio,BottomRatio) TopHalfAreaMineral = sum(areamineral_array(1:round(end/2)))*(Resolution^2) BottomHalfAreaMineral = sum(areamineral_array(round(end/2):end))*(Resolution^2) handles.finalcontourdarea = contourarea_finalmm; handles.finalconnectedarea = connectedarea_finalmm; guidata(hObject,handles) % set(handles.TotalVolumeText, 'String',['Contour Area: ' num2str(contourarea_finalmm) ' mm^2']); % set(handles.NormalizedVolumeText, 'String',['Connected Area: ' num2str(connectedarea_finalmm) ' mm^2']); set(handles.TotalVolumeText, 'String',['See Matlab Window for Area Measures']); set(handles.NormalizedVolumeText, 'String',[' ']); data(2,3:18) = {sprintf('%0.4f', contourarea_NoTopCapmm),sprintf('%0.4f', connectedarea_NoTopCapmm),... sprintf('%0.4f', TopHalfContourArea), sprintf('%0.4f', TopHalfConnectedArea), sprintf('%0.4f', BottomHalfContourArea), sprintf('%0.4f', BottomHalfConnectedArea), sprintf('%0.4f', WholeContourArea), sprintf('%0.4f', WholeConnectedArea),... sprintf('%0.4f', TopRatio), sprintf('%0.4f', BottomRatio), ... sprintf('%0.4f', MinimumConnectedRatio), sprintf('%0.4f', mingray), ... sprintf('%0.4f', TopHalfAreaMineral), sprintf('%0.4f', BottomHalfAreaMineral), sprintf('%0.4f', WholeAreaMineral), sprintf('%0.4f', WholeAreaMineral/WholeConnectedArea)}; pathname = handles.pathname; [file, path] = uiputfile('*.xls', 'Save Excel File', pathname); xlswrite([path file], data, 'sheet1'); pause(0.01) close(h) pause(0.01) beep on pause(0.5) beep off end % - - - - - - - - - - - - - - - - - - - - - % use this script to light the 3D surface rendering % of the output of the connectivity by surface area % I.e. adds lighting to figure of UnionRatio function UASurfaceLightingScript() % save original figure first!! global lighthandle; if isempty(lighthandle) lighthandle = camlight; end axis off lighting phong j = colormap(jet); j(1:end, 1:end) = 0; j(1,3) = 0.5; j(2:end,1) = 0.9; colormap(j); lighthandle = camlight(lighthandle, 'headlight'); ka=0.3; kd=0.7; ks=0.5; n=15; sc=1; material dull % material([ka kd ks n sc]) ; % sets the ambient/diffuse/specular strength, % specular exponent, and spec ular color reflectance of the objects. end

169

7.2 APPENDIX B: PTH FOR FRACTURE HEALING LITERATURE REVIEW

Authors  Year   Animal Model / Age 

Injury Model 

PTH Treatment Other Treatments/Variables 

Major Findings

Andreen  1983  Sprague‐Dawley Rats (male) 

Closed tibial fractures  

Parathyroidectomy   vitamin D for rescue of PTX? 

Parathyroidectomy  impairs fracture healing and resulted in non‐unions, Vitamin D could not rescue it much. 

Centrella  1989           PTH modulates TGF‐b activity and binding in osteoblast enriched cell cultures 

Fang  1996  Rats  Critical osteotomy filled with GAM

PTH(1‐34) plasmid + BMP‐4 plasmid 

  Combination therapy of BMP‐4 and PTH gene increased bone formation greater than BMP‐4 alone 

Andreassen   1999  Female Wistar Rats 

Closed tibial fractures  

60 μg/kg PTH or 200 μg/kg PTH for 20 or 40 days 

  at 20 days, only 200μg/kg PTH enhanced fracture healing strength (to 14% of contralat.) with larger callus of higher BMD (DEXA)

      3 months old 

      at 40 days, both significantly enhanced fx strength (to 47% of contralat.) 

               contralat tibia sig. 15% stronger after 40 days of 200μg/kg

Kim  1999  Ovariectomized Rats 

Bilateral tibial shaft fractures 

PTH(1‐84) 17‐estradiol 

PTH improved morphometric and mechanical parameters dose dependently 

               17‐estradiol (bone‐resorption inhibiting agent) did not benefit healing 

170

Bostrom  2000  Male New Zealand White Rabbits 

1mm osteotomy in ulna 

PTHrP analog Prednisone corticosteroid  

PTH reversed the inhibition of bone healing by prednisone to improve radiographic union rate and mechanical strength and stiffness 

Skripitz  2000  JBJS 

Male Sprague‐Dawley Rats 350g 

Bone Chamber in proximal tibial metaphysis

0, 15, 60 or 240μg/kg PTH 

sacrificed at 6 weeks 

all doses of PTH enhanced cancellous density in the chamber, which correlated linearly with the log([PTH]) dose 

               distance of bone ingrowth into the chamber was unchanged 

Skripitz  2000 Acta Orthop. 

Male Rats 350g 

Bone chamber in proximal tibial metaphysis   

60μg/kg PTH time sequence studies with endpoints at 2, 4, 6 weeks  

chamber cancellous density decreased in control animals, while PTH treated density increased over time 

           slight increase in bone ingrowth depth

Andreassen   2001  Old Wistar Rats 

Tibial fracture 

200 μg/kg PTH for 3 or 8 weeks 

  at 21 and 56 days, the fractures were sig. stronger with more mineral and greater girth bu pQCT

      27 months old (2.25 yr) 

      56 day fx tibia were >2x stronger than contralateral! 

Neer   2001  Women Postmenopaus 

  placebo, 20, or 40 μg of PTH 

  rate of new vertebral fxs was 14% (64 of 448), 5% (21 of 444) and 4% (20 of 434) respectively

               relative risk of fracture to placebo were 35% and 31%

171

               non‐vertebral fxs rates were 6%, 3%, 3% respectively

               BMD (DEXA) increased by 9% (20μg/kg)and 13% (40μg/kg) over placebo in the vertebrae 

               BMD (DEXA) increased by 9% and 13% over placebo in the vertebrae and by 3% and 6% in femoral neck, and 2% and 4% in whole body 

Skripitz  2001  Male Rats Steel screw implant into proximal tibia

60μg/kg PTH   implant fixation strength and bone fraction were significantly greater at 2 and 4 wks 

Nakajima   2002  Sprague‐Dawley Rats 

Closed femoral fractures

2, 10, 50, 100 μg/kg PTH 

  Minimum effective dose for fracture healing: 10μg/kg 

      2 months old 

  10μg/kg PTH   PTH stimulated osteoprogenitors in periosteum at the fx site at day 2, but not later

               PTH  increased osteoclast index at day 7, but not later

               increased gene expression of marker for differentiated osteoblast 

               IGF‐1 expression levels greater in PTH treated animals

Vahle  2002  Fisher 344 Rats  

  5, 30 and 75 μg/kg PTH

  all doses caused substantial increase in bone mineral

      6‐8 weeks, given for 2 years 

no injury     Trabecular Hypertrophy, filling of intramedullary space, increased extramedullary hematopoesis (enlarged spleen, liver) 

172

               All bone proliferation lesions (osteosarcoma, osteoblastomas) observed were in PTH treated animals

               (no placebo treated animals had cancer)Chen  2003  Adultmale 

Sprague‐Dawley Rats 450 ‐ 550g 

critical femure osteotomy filled with gene activated matrix

Local PTH plasmid by GAM plus systemic PTH injected 

  combination therapy improved the bone mineral in the 5mm osteotomy gap over systemic PTH. 

Okazaki  2003  Rats  fracture     PTH/IGF‐1 interaction                Zhuo  2003  C57Bl/6 

Mice Ovariectomy 

40μg/kg PTH or vehicle started 4 weeks after OVX 

ovariectomy  

normal mice PTH induced bone volume changes: Vertebrae > proximal tibia trabec >> tibia diaphysis 

      10 wk females 

sham or after after sham surgery 

  OVX mice +/‐ PTH: OVX caused slight reduction in prox tib BVF which was not affected by PTH.  OVX caused reduction in vertebral BVF which was sig improved with PTH.  No change in tibia diaphysis due to OVX, slightly improved with PTH treat.

               comparing axial and apendicular bone loss and formation rate due to PTH treatment suggested that the number of bone surface areas in a given region explained these changes in BVF  

173

Andreassen  2004  Female Wistar Rats 

tibial fracture 

intermittant Systemic PTH 8 wks after Fx followed by 8 wks of withdrawl     

  PTH increases fracture strength by 8 weeks.  After 16 weeks, there is no difference in mechanical props. 

      3 months old 

    Callus strength continues to increase during the next two weeks even after withdrawl of treatment of PTH 

             at 16 weeks, treated and untreated fracture mechanics are equal 

Seebach  2004  Sprauge‐Dawley Rats 

Distraction osteogenesis of the femur

60μg/kg PTH   PTH increased strength, stiffness, callus volume, callus BMC and density relative to untreated.   

      3 months old 

      Contralateral femur also slightly stronger

                  PTH could be useful to shorten the distraction osteogenesis consolidation time. 

Alkhiary  2005  Sprauge‐Dawley Rats 

closed femur fracture 

5μg/kg and 30μg/kg PTH (1‐34) for up to 35 dayds 

  30μg/kg increased bone content and density and percent of cartilage in callus, resulting in greater max torque and stiffness slightly by 21 days 

      450g        5μg/kg increased the callus bone content and mechanics 

               higher strength and bone mineral content was maintained at day 84 even after withdrawl of 30μg/kg since day 35 

174

               no increase in osteoclast density in callus (actually, slightly decreased) 

Komatsubara 

2005  Female Sprauge‐Dawley Rats 

osteotomy at 8 weeks old 

PTH 10 μg/kg or 30 μg/kg initiated 3 wks before osteotomy

after fx, PTH treatment was either withdrawn or continued should PTH treatment for Osteoporosis be stopped or continued after fx?  

animals receiving PTH 3 wks before osteotomy were better off if they continued to received PTH, especially at  30μg/kg 

      5 weeks old 

    New bone formation and the conversion from woven to lamellar bone was improved with PTH treatment after osteotomy, and the ultimate load in 3 point bending was greater by 12 weeks with 30μg/kg but not before, and not with 1μg/kg

             there is no reason to terminate PTH treatment when a fx occurs, infact it's better to maintain treatment 

Nakazawa  2005  Sprague‐Dawley Rats 

Femur fracture 

10μg/kg PTH (1‐34) given to half of fractured animals 

  PTH enhanced cartilage area in Fx at day 14 (but not at day 7, 21, 28) 

      2 month old 

      PCNA (proliferating cell nuclear antigen) staining showed PTH stimulated mesenchymal area of fx. At day 4 & 7, but in the cartilage area of fx. ‐ suggesting PTH stimulates stem cells greater than chondrocytes

               Sox9 and Col 2 upregulated a little

175

             also looked at High dose PTH on growth plate for 2 weeks 

Slight, insignificant change in growthplate length with 20μg/kg. GP hypertrophic zone length was significantly greater after 2wks of 400μg/kg, and 800μg/kg.  Administering 800μg/kg does not yield more growth plate than 400 .

Pettway  2005  C57Bl/6 BMSC donors implanted into Nude mice 

subcutaneous BMSCs on gelatin sponges 

harvested after 1, 3, and 7 wks of PTH 30μg/kg treatment 

also studied gene expression after transient (8hr) PTH treatment on day 7 and 14   

ossicles from mice treated for 3 weeks had much greater mineralization, but by 7 weeks they were equivalent to ossicles in untreated mice, suggesting the ossicles the effect of PTH on BMSCs is temporally dependent probably due to differentiation stage and that they are particularly responsive to PTH for the 1st 4 weeks

      4‐8 weeks old 

    3 weeks of PTH treatment initiated 12 weeks after implantation only stimulated a small increase in mineral content. 

Aleksyniene  2006  Female New Zealand White Rabbits 

Distraction osteogenesis of the tibia 

5 or 25μg/kg PTH   Callus size, BMD, BMC, trabecular measures was increased dose dependently with PTH treatment 

      6‐8 months old 

      25 μg/kg PTH was significantly more effective than 5 microg/kg/day PTH(1‐34) 

Gabet  2006  Male Sprague‐Dawley Rats 

Threaded titanium implant in metaphysis 

5, 25 or 75 μg/kg/day 

Ti rods were implanted 7 weeks postorchie

25 and 75μg/kg PTH improved implant fixation after 8 weeks  

176

ctomy 

      5 months old 

      PTH improved trabecular bone structural indicies and the surface osteointegration was improved

Abe  2007  Male Sprague‐Dawley Rats 

Spinal fusion with morselized autograft

40μg/kg PTH   spinal fusion rate was greater with PTH treatment by 14 days, fusion mass was larger and denser at 14, 28 and 42 days 

      8 weeks old 

      PTH stimulated bone formation and bone resorption gene expression in the fusion mass

               number of osteoclasts in fusion mass was increased with PTH treatment at D24 and 42 

Gardner  2007  Male C57Bl/6 Mice 

open osteotomy 

30μg/kg PTH treated on days 5 ‐ 18, then sac'd) 

daily, cyclic, compressive loading (on days 5‐18, then sac'd) 

PTH induced osteoblast osteoid production

      10 weeks old 

      Loading induced osteoclastic activity 

               PTH+Load lead to higher callus BMD and BVF and synergy was suggested as relavent, but PTH alone improved bending strength 

177

Hashimoto  2007  Male Lewis Rats 

Live vascularized isogenetic allograft for arthrodesis     

10 or 100 μg/kg PTH 

followed by 2μg/kg Zolendronic Acid for 4 weeks   

PTH increased serum bone formation markers, ZA supressed both resorption and formation markers. 

      12 wks old   4 wks PTH followed by vehicle improved grafted bone strength but not as much as 8wks of PTH treatment, or 4wks PTH followed by ZA for 4wks which were relatively equivalent

             conclusion: treatment with ZA following PTH treatment maintains bone density and strength equally well as continued PTH treatment

Kakar  2007  Male C57Bl/6 Mice 

Closed femoral fractures

30μg/kg PTH (1‐34) for 14 days 

  PTH enhanced fracture callus volume at D14, especially cartilaginous volume 

      8 weeks old 

      PTH enhances chondrocyte genes and cause them to peak earlier (Sox9) to a slightly greater extent than the osteogenic genes (RunX2, Osterix) 

               PTH enhanced chondrocyte hypertrophy (Sox5 and ColX)

               PTH enhanced Wnt 5a expression in callus from days 2 ‐7 

               PTHrP and IHH signalling are also enhanced in the callus

               any increase in B‐Catenin levels were not conclusive

178

Manabe  2007  Cynomolgus Monkeys Female 

femoral osteotomy 

0.75μg/kg or 7.5μg/kg 2x/wk initiated 3 weeks before osteotomy 

not sacrificed until after 6 months 

Hi dose PTH improved material strength and modulus but had no effect on whole bone mechanics 

      18 or 19 years old 

      Callus area of PTH treated was slightly less, %bone area in callus was significantly greater 

               Osteoclast number within the callus dose was reduced dose dependently with increasing PTH

               Hi PTH increased mineral aposition rate and activation frequency 

Rozen  2007  Female Wistar Rats 

tibial fracture 

1ug, Local PTH(1‐34) on day 4,5,6 post fx  

followed by local IL‐6 + IL‐6 soluble receptor on day 7, 9, 11 with 3 different doses       

callus volume: control < IL‐6 < PTH(1‐34) == PTH(28‐48) < PTH(1‐34)+IL6+R == PTH(28‐48)+IL6+R 

      200 ‐ 250g   or 0.2ug or 1ug local PKC‐specific PTH(28‐48), day 4, 5, 6 

mechanical strength: control == PTH(1‐34) < PTH(28‐48) < PTH(28‐48)+IL6+R < PTH(1‐34) + IL6+R 

             PTH during cartilage anlage formation induced cartilage

             activation of osteoclasts by IL‐6 accelerates callus remodelling 

Trisidis  2007  New Zealand white rabbits 

Metaphyseal wedge osteotomy filled with b‐

systemic PTH 10μg/kg, vs 40μg/kg vs local 200ng OP‐1 

  Only local OP‐1 improved torsional strength, stiffness and bone mineral content 

179

      16 weeks old   

TCP  

   

  Low dose PTH had the greatest increase in BMC but did not improve strength.   

          hi dose PTH had little effect on BMC or mechanics

Aspenberg  2008  Sprague‐Dawley rats 295‐481g 

Screw insertion into the tibia 

Daily Injections of PTH 

Screw coating with Bisphosphonates

The combined treatment signficantly increased screw pullout force and energy 

      11 weeks old 

  (Unkown dose) Pamidronate (300 ng/cm2) and Ibandronate (340 ng/cm2) 

PTH+Bisphos > PTH > Bisphos > Untreated Controls 

Iwaniec  2008  Male Sprague‐Dawley Rats 

Demineralized bone matrix ectopic bone formation cylinders

1μg/kg PTH (very low!) 

Chronic alcohol abuse 

Alcohol consumption decreased whole body BMC, PTH enhanced whole body BMC, but not on periosteal bone nor within the chamber.  Alcohol impaired bone formation and reduced the benefits of PTH treatment 

      3 months old 

sac'd at 6 wks

    BMC:  Alcohol < Control == PTH+Alcohol < PTH

Johansson  2008  Sprague‐Dawley rats 

steel screws and steel rod implanted into the 

60μg/kg PTH withdrawl of PTH after 2 weeks of treatment, 

PTH treated animals had greater implant pullout strength up to 8 days after PTH withdrawl, afterwhich there was no difference 

180

tibial metaphysis 

or replaced with 500μg/kg Pamidronate

      12 weeks old 

      At 3 weeks after PTH withdrawl, Pamidronate treatment maintained twice the pull out strength of animals with 2wks of PTH followed by no treatment.

Kaback  2008  C57BL/6 mice 

closed femoral fractures 

40μg/kg PTH cell culture  marrow MSCs from mice treated with PTH were more osteoblastic probably through induction of Osterix and Runx2 expression 

      7‐9 weeks old 

      Osterix and RunX2 genes were upregulated in the fractures of PTH treated aminals along with early Col2 and Col1 

Nozaka  2008  Female Wistar rats 

osteotomy 100μg/kg PTH once per week 

+/‐ OVX  PTH increased cancellous bone volume by stimulating bone formation in both normal and OVX rats and suppressed adipocyte marrow volume

      7 months old 

      PCNA Proliferating cell number was 2‐3 fold greater with PTH treatment, especially at the osteotomy site

Pettway  2008                                            Morgan  2008  New 

Zealand white rabbits 

Metaphyseal wedge osteotomy filled with b‐

Half of them got 10μg/kg PTH 

200μ into half the b‐TCP scaffolds to 

rhBMP7 had little effect on bone formtion, torsional or compressive strength with or without PTH treatment.  PTH+BMP7 was not sig. greater than PTH alone.

181

TCP study the combination 

                Vahle  2008  Cynomolg

us Monkeys Female (OVX) 

no injury half got 5ug/kg PTH for 18 months 

studied for 18 months and 4.5 years 

18 months of PTH caused no neoplasms (observed for 3 years after treatment also) 

               PTH improved BMD in vertebrae and femoral neck after 18 months of treatment, but the effect in the vertebrae was lost after 3 years, while the femoral neck retained increased BMD due to PTH even after 3 years after treatment termination.

182