Top Banner
Copyright by Taneidra Walker Buie 2020
210

Copyright by Taneidra Walker Buie 2020 - Front Matter Template

Apr 29, 2023

Download

Documents

Khang Minh
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

Copyright

by

Taneidra Walker Buie

2020

Page 2: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

The Dissertation Committee for Taneidra Walker Buie Certifies that this is the

approved version of the following Dissertation:

DEVELOPMENT OF MULTIFUNCTIONAL ELECTROSPUN

WRAPS FOR BONE HEALING

Committee:

Elizabeth Cosgriff-Hernandez, Supervisor

Laura Suggs

Janeta Zoldan

David Laverty

Page 3: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

DEVELOPMENT OF MULTIFUNCTIONAL ELECTROSPUN

WRAPS FOR BONE HEALING

by

Taneidra Walker Buie

Dissertation Presented to the Faculty of the Graduate School of

The University of Texas at Austin

in Partial Fulfillment

of the Requirements

for the Degree of

Doctor of Philosophy

The University of Texas at Austin

December 2020

Page 4: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

Dedication

To my dear ancestors, who fought to make life better for me.

I am your wildest dreams.

Page 5: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

v

Acknowledgements

This journey, by far, has been the most challenging, yet rewarding, journey that I

have ever endured. The growth that I have experienced both personally and professionally

would not have been possible without several key mentors, collaborators, peers, family

members, and friends.

First, I would like to give thanks to God. I know that it is not conventional for

science, religion, and spirituality to mix. Even frowned upon by some. However, nothing

in this world could stop me from giving You the praise and worship that You so deserve.

You made it my destiny to become something bigger than myself. Graduate school was the

vision that You implanted in me to bring this destiny to fruition. My faith in You and Your

vision of my life is what granted me the strength to see it through. So, thank you, thank

you, and thank you for showing me what is in store for me if I trust in Your leadership. It

was and will always be worth it.

Next, I would like to thank my advisor, Dr. Elizabeth Cosgriff-Hernandez. Without

you believing in me and consistently pushing me, I would have never realized the potential

that I harness as an independent researcher, a mentor, and a leader. You have instilled in

me that I am capable of achieving anything as long as I apply myself. It is your commitment

to my training that has allowed me to become the best professional version of myself that

I am today.

Page 6: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

vi

My dissertation would not have had as great of an impact in the field if it were not

for my committee, Dr. Laura Suggs, Dr. Janet Zoldan, and Dr. David Laverty. Thank you

for your guidance and mentorship in bringing forward the pivotal change in direction that

was needed to advance this work. In addition to my committee, I would like to thank my

collaborators, Dr. Joseph Wenke and Dr. Michael Whitely for your assistance in advancing

the direction of the antimicrobial work and for providing valuable feedback whenever I

needed it. To Dr. Noah Cohen and Dr. Canaan Whitfield-Cargile, your help with designing

and evaluating the anastomosis bilayer wraps is greatly appreciated. Also, working with

you both provided me with one of many exciting moments in graduate school- developing

and observing dopamine-adhesive meshes stick to various tissues. So, thank you for that.

Another cherished moment was working with Jacob Blacutt (Dr. Vernita Gordon’s lab).

Your guidance and expertise, along with Dr. Gordon’s generosity, allowed me to develop

a microbial culture set-up and relevant protocols for our lab that will be used for various

future projects. Similarly, I would like to thank Austin Veith (Dr. Aaron Baker’s lab) for

training me, assisting me, and providing me with the supplies to perform my first animal

study- by far the highlight of my graduate journey.

Although I am completing my graduate journey at the University of Texas at

Austin, I did not start here. I would not have had a successful dissertation and an overall

amazing graduate experience had I not started with a strong foundation at Texas A&M

University. I was on the brink of canceling my plans to advance my degree and moving

back home to comfort, but a group of mentors and friends swooped in and supported me

during those tough times. A special thank you to Shawaneé Patrick, Dr. Samuel

Page 7: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

vii

Merriweather, and Dr. Shannon Walton for always giving me guidance that often felt like

it was coming directly from a wise family member. Thank you to my Black Graduate

Student Association crew for giving me a space that felt familiar with like-minded people.

The mentorships and friendships that I established there definitely shaped my overall

outlook on my graduate experience. For the first time ever, I saw Black excellence in

STEM in the form of several influential leaders. I aspired to be like you all, and the only

way I saw that I could make that happen was to stay the course. So, thank you for

unknowingly influencing me to stay.

My dissertation would not have been possible if not for the funding sources that I

have received. I would like to acknowledge the National Science Foundation (Texas A&M

University Bridge-to-the-doctorate Program and Graduate Research Fellowship Program)

and the National Institutes of Health (R03 AI136060). I would also like to acknowledge

The University of Texas at Austin for their financial support through generous fellowships

and scholarships (2020 Agnes T. and Charles F. Wiebusch Fellowship and 2019

Engineering Foundation Endowed Graduate Presidential Scholarship) as well as for

funding me after completion of my external fellowships.

One of the most rewarding experiences during my time in graduate school was the

ability to work with several talented lab members. Although you may not read this, I have

to start by saying thank you, Dr. Alysha Kishan. Your foundational work paved the way

for me to achieve all that I have in this lab. I can only hope that I leave a legacy as great as

yours for the next generation in our lab. To my current lab members, you all have made

this journey much easier. I have grown to have a special bond with you all and will miss

Page 8: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

viii

you all dearly. To my dearest Prachi Dhavalikar, I will miss you the most. We grew to be

more than lab members and more than friends. You and I have developed a bond over the

last five years that is indescribable and that extends beyond the confines of our academic

journey. I could not have imagined this journey without you, and I will forever cherish

your support, kindness, and friendship. You played a pivotal role in me finding one of my

greatest purposes- helping others to realize their potential. As you travel through life, I

hope that you hold on to the advice and words of encouragement that I have given you. I

would also like to give a special thanks to all of the chemistry gurus, Gabriel Rodriguez-

Rivera, Megan Wancura, and Dr. Malgorzata Chwatko. You all were always so willing to

help me feel a little less incompetent in this area. To the cell and microbial culture gurus,

Prachi Dhavalikar, Dana Jenkins and Ziyang Lan, thank you for helping me brainstorm

through different ideas for these studies throughout the years. To the electrospinning gurus,

Andrew Robinson and Sarah Jones, I know that our time together has been the shortest, but

I hope that you were able to learn from me as I have already learned so much from you

both. Aside from the technical aspects, I truly appreciated and will miss our camaraderie,

our unconditional personal and professional support of one another, our lunch dates at

Madam Mams (but not Taco Joint), and our much-needed happy hours. I could not have

asked for a more supportive group of people to work during graduate school.

I also could not have done most of this work without the amazing army of

undergraduate students with whom I had the pleasure of mentoring and training. To Joshua

McCune, Anupriya Jose, Sophia Ty, and Annika Balakrishnan, thank you for your hard

work, your persistence, and your desire to learn. You all were the heart of this research.

Page 9: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

ix

You all treated this work as if it were your own dissertation, and that dedication is what

allowed us to make so much progress in our short time together. Working with you all

taught me more about myself than you could imagine. I grew to love mentoring because

you all made it so fun and worthwhile. I wish you all well on your next journey and can

only hope that our time together impacted your lives in the most meaningful way. Be great

in all that you do my little minions!

Last, but certainly not least, I am indebted to my loving and supportive family and

friends. Graduate school is full of many ups and downs, but you all were there to cheer me

on through all of them. To my husband, James (Jamie) Buie, thank you for weathering the

storm with me. Jamie, I know that this journey was not easy for you and that it took a toll

on our marriage. I recognize that you sacrificed a lot ‒ your hobbies, your friends, your

career‒ just to be by my side and stand in support of my goal. For that, I am eternally

grateful. Although I did not frequently say this, I could not have managed this journey

without you. I truly believe that we are coming out of this experience together much

stronger than we started. We can finally exhale. I love you. To my parents, Letitia and

Laferrell Walker, and my sister, Feaundra (Fee) Walker, I thank you for your unconditional

love, support, and encouragement. Daddy, without your tough love, life advice, and superb

negotiation skills, I would have not initially taken the risk to embark on this journey.

Mama, without your comforting words, daily conversations, and random care packages, I

would not have had the courage to stay away from everything that I have ever known and

loved just to achieve my goal. Fee, you often tell me that I am your role model. Those

words are what kept me on course when I wanted to give up many times on this journey.

Page 10: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

x

Without you all backing my vision from God, graduate school would not have been a reality

for me. To my fur-baby, Coal, thank you for providing unspoken comfort during my most

stressful seasons during graduate school. You will never know the impact you had on me

and my sanity. You gave me unconditional love and licks that were always sure to brighten

any dark day. To my grandparents, Minnie and Frank Wilson and Julius Walker, as well

as a host of aunts, uncles, cousins, in-laws, and best friends, thank you for reminding me

of my perseverance, willfulness, and fortitude that has always guided me through

challenging times. These reminders grounded me when things started to seem impossible.

I initially started this journey to become a role model for you all; to become something

greater than myself. I wanted to be an example of the greatness that comes from our roots.

I hope that I have inspired you all to challenge yourselves and step out of your comfort

zones. Greatness awaits you too!

Page 11: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

xi

Abstract

Development of Multifunctional Electrospun Wraps for Bone Healing

Taneidra Walker Buie, Ph.D.

The University of Texas at Austin, 2020

Supervisor: Elizabeth Cosgriff-Hernandez

The Masquelet technique is a two-staged procedure that uses an induced biological

membrane and bone graft to reconstruct critical-sized bone defects. However,

unpredictable clinical outcomes result due to the variable durability and the transient

vascular network of the induced membrane, as well as high incidences of osteomyelitis. To

this end, we have engineered a resorbable multifunctional electrospun wrap that guides

formation of the induced membrane with improved durability and enhanced angiogenesis

while simultaneously preventing infection. We achieve this by developing and combining

an antimicrobial poly(lactic-co-glycolic) acid (PLGA) mesh and an angiogenic crosslinked

gelatin mesh.

We first confirmed the ability of electrospun PLGA to provide sustained release of

gentamicin sulfate or gallium maltolate above its minimum inhibitory concentration

(MIC). Studies that evaluated antimicrobial activity indicated that osteomyelitis-derived

bacteria was not susceptible to released gallium maltolate at the hypothesized MIC and

Page 12: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

xii

further established the accurate gallium maltolate MIC. The inhibitory concentration of

each antimicrobial on osteoblasts was compared to the respective MIC to determine if they

were safe and effective at released concentrations. Results concluded that the gentamicin

sulfate-loaded PLGA mesh is safer and more effective mesh. Next, the bioactivity retention

of vascular endothelial growth factor (VEGF) released from electrospun photo-crosslinked

gelatin-methacrylate was confirmed. Subcutaneous implantation of the VEGF-loaded

mesh in a rat corroborated resorption and the capacity for sustained release. A

multifunctional electrospun wrap was then engineered to prevent osteomyelitis and guide

formation of the induced membrane by combining the antimicrobial and angiogenic

platforms with co-electrospinning. The combination of the two fiber populations was

confirmed microscopically and offered independently tuned bimodal release of gentamicin

sulfate and VEGF.

Overall, this work provides the fundamentals to advance the development of a

multifunctional electrospun wrap that can guide formation of the induced membrane and

prevent osteomyelitis for improved clinical outcomes with the Masquelet technique. This

work offers a substrate that can recruit and support cellular adhesion, provide a template

for matrix deposition and tissue remodeling, and enable bimodal release of bioactive

agents. These studies also enhance the capacity of electrospun platforms to serve as stand-

alone therapies or combinatorial therapies in various bone regeneration applications.

Page 13: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

xiii

Table of Contents

Dedication .......................................................................................................................... iv

Acknowledgements ..............................................................................................................v

Abstract .............................................................................................................................. xi

List of Tables ................................................................................................................... xvi

List of Figures ................................................................................................................. xvii

Chapter I: A Critical Review of Biomaterial Approaches for Improved Bone

Regeneration with the Masquelet Technique .......................................................................1

1.1 Bone Loss Management........................................................................................1

1.2 Biological Role of the Induced Membrane ...........................................................3

1.2.1 Characterization of the Induced Membrane ..................................3

1.2.2 Limitations of the Induced Membrane ..........................................4

1.2.3 Recent Approaches to Guide Membrane Formation ....................6

1.3 Improve Mechanical Durability ............................................................................7

1.3.1 Freeze-drying ................................................................................8

1.3.2 Microfluidic Spinning ...................................................................9

1.3.3 Electrospinning ...........................................................................10

1.3.4 Fibrous Scaffolds to Improve Durability ....................................11

1.4 Enhance Vascularization.....................................................................................12

1.4.1 Delivery of Angiogenic Factors ..................................................13

1.4.2 Gene Delivery .............................................................................16

1.4.3 Integrin Targeting .......................................................................17

1.4.4 Cell Delivery ...............................................................................19

1.4.5 Mechanisms to Enhance Vascularization ...................................20

Page 14: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

xiv

1.5 Prevent Osteomyelitis .........................................................................................21

1.5.1 Antibiotics ...................................................................................22

1.5.2 Metals ..........................................................................................24

1.5.3 Antimicrobial Peptides................................................................26

1.5.4 Resorbable Matrices for Antimicrobial Delivery .......................28

1.5.5 Local Delivery of Antimicrobials ...............................................30

1.6 Summary and Approach .....................................................................................30

Chapter II: Comparative Efficacy of Resorbable Fiber Wraps Loaded with

Gentamicin Sulfate or Gallium Maltolate in the Treatment of Osteomyelitis ...................33

2.1 Introduction .........................................................................................................33

2.2 Materials and Methods........................................................................................37

2.3 Results .................................................................................................................46

2.4 Discussion ...........................................................................................................54

2.5 Conclusions .........................................................................................................60

Chapter III: Gelatin Matrices for Growth Factor Sequestration .......................................61

3.1 Polymeric Matrices for Growth Factor Delivery ................................................61

3.2 Affinity Sequestration to Control Growth Factor Release..................................63

3.3 Gelatin Matrices in Tissue Engineering..............................................................71

3.3.1 Gelatin Microparticles ................................................................72

3.3.2 Gelatin Scaffolds .........................................................................75

3.4 Future Perspectives in the Masquelet Technique ...............................................78

Chapter IV: A Multifaceted Matrix to Enhance Angiogenesis and Provide Infection

Control during Bone Regeneration ....................................................................................82

4.1 Introduction .........................................................................................................82

Page 15: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

xv

4.2. Materials and Methods.......................................................................................85

4.3 Results .................................................................................................................95

4.4 Discussion .........................................................................................................106

4.5 Conclusion ........................................................................................................113

Chapter V: Conclusion .....................................................................................................115

5.1 Summary ...........................................................................................................115

5.2 Significance of Work ........................................................................................117

5.3 Challenges and Future Perspective ...................................................................121

Appendix A: In Vivo Performance of a Bilayer Wrap to Prevent Abdominal

Adhesions ........................................................................................................................126

A.1 Introduction ......................................................................................................126

A2. Materials and Methods .....................................................................................129

A.3 Results ..............................................................................................................138

A.4 Discussion ........................................................................................................149

A.5 Conclusions ......................................................................................................155

References ........................................................................................................................157

Page 16: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

xvi

List of Tables

Table 1.1: Summary of angiogenic factors indicated to regulate angiogenesis. ...............14

Table 1.2: Summary of ligands commonly used for integrin targeting of vascular

endothelial cells. ..............................................................................................18

Table 1.3: Summary of the common classes of antibiotics indicated to treat bone

infection. ..........................................................................................................22

Table 1.4: Summary of metallic antimicrobials indicated to prevent bone infection. ......24

Table 1.5: Proposed structure-activity relationship of various antimicrobial peptides. ....26

Table A.1: Tensile properties of composite bilayer wrap formulations. * indicates

statistical differences as compared to the hydrogel foam-fiber composite,

(P<0.05). .......................................................................................................141

Table A.2: Adhesion scoring description. .......................................................................146

Page 17: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

xvii

List of Figures

Figure 2.1: Fabrication of antimicrobial wraps. A) Schematic of electrospinning

apparatus and scanning electron micrographs of electrospun PLGA fibers

loaded with B) gentamicin sulfate and C) gallium maltolate. Main

micrograph scare bar = 100 µm. Inset micrograph scare bar= 30 µm. ..........47

Figure 2.2: Evaluation of in vitro release kinetics from electrospun antimicrobial

PLGA wraps in DI water. Cumulative release and daily release of A)

gentamicin sulfate and B) gallium maltolate was evaluated over 2 weeks.

Red-dashed line indicates hypothesized MIC for each antimicrobial. ...........49

Figure 2.3: Kirby Bauer assay was used to evaluate bioactivity of the antimicrobial

wraps. Representative images of the inhibition zones in response to A)

gentamicin sulfate and B) gallium maltolate released from the PLGA

wrap (bottom half images), as compared to negative control (blank PLGA

wrap, top right image) and the positive control (solubilized gentamicin

sulfate or gallium maltolate, top left image) after 24 h. Graphs display the

corresponding measurements of zone diameters. * indicates statistical

differences with respect to the gentamicin sulfate positive control

(P<0.05). .........................................................................................................51

Page 18: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

xviii

Figure 2.4: The evaluation of the MIC and MBC of A) solubilized gentamicin sulfate

and B) solubilized gallium maltolate on MSSA bacterial colony growth

after 24 h. The mean initial bacteria density is denoted by the red dashed-

line. The MIC was deemed the lowest concentration that inhibited

bacterial growth such that the treated bacteria density (CFU/mL) is not

statistically different than the initial bacteria density. * indicates

statistical differences with respect to the initial bacteria density, (P<0.05).

The blue dashed-line represents a 99.9% reduction in the initial bacteria

density. The MBC was deemed the lowest concentration that reduced

bacterial growth ≥99.9% of the initial bacteria density. .................................53

Figure 2.5: Viability of differentiated MC3T3-E1 cells relative to TCPS control after

24 h exposure to various concentrations of A) solubilized gentamicin

sulfate and B) solubilized gallium maltolate. Red-dashed line indicates

relative IC50 calculated using GraphPad Prism 8. A relative IC50 was

identified as 566.5 ± 142.4 µM for gentamicin sulfate and 778.6 ± 326.1

µM for gallium maltolate. ..............................................................................54

Figure 3.1: The degree of crosslinking affects the hydrogel mesh size that governs

growth factor release from gelatin matrices. A) Low crosslinking results

in rapid swelling and diffusion. B) High crosslinking results in reduced

swelling and sustained diffusion. ...................................................................65

Figure 3.2: Effect of conjugation on growth factor sequestration in gelatin matrices.

Growth factor-conjugated gelatin matrix displays burst release due to

initial swelling that releases non-conjugated growth factors followed by

sustained growth factor release after proteolytic chain scission. ...................66

Page 19: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

xix

Figure 3.3: Overview of the physiochemical properties governing growth factor

diffusion from gelatin matrices. The properties included growth factor

affinity to A) ligands, B) adaptor proteins, and C) nanomaterial additives

incorporated into gelatin matrices. .................................................................67

Figure 3.4: Effect of construct surface area-to-volume ratio on growth factor

diffusion from gelatin microparticles. A) Smaller microparticles have

shorter diffusion path lengths leading to rapid release of growth factors;

B) larger microparticles have longer diffusion path lengths and slower

release profiles. Scanning electrospun micrographs reprinted with

permission. .....................................................................................................74

Figure 3.5: Effect of construct surface area-to-volume ratio on growth factor

diffusion from gelatin fibers. The diffusion path length in electrospun

constructs are controlled by fiber diameter with A) thin fibers having

shorter diffusion path lengths and rapid release; B) thick fibers have

longer diffusion path lengths and slower release profiles. Representative

scanning electron micrographs reprinted with permission from [251]. .........77

Figure 3.6: Bimodal release of model proteins (FITC-bovine serum albumin and

TRITC-bovine serum albumin) from a single electrospun gelatin-based

mesh in collagenase. Figure reprinted with permission from. ......................80

Figure 4.1: NMR spectra of A) gelatin and B) gel-MA used to quantify

functionalization. ............................................................................................96

Page 20: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

xx

Figure 4.2: Capillary-like network formation in response to unprocessed VEGF. A)

Representative images of network formation induced by increasing

concentrations of unprocessed VEGF. Cells stained with calcein-AM.

Scale bar is 200 µm. B) Quantified network formation/field of view

corresponding to the representative images. ..................................................98

Figure 4.3: Evaluation of the bioactivity of released VEGF from electrospun gel-MA

meshes. Representative images show capillary-like network formation

corresponding to blank releasate and releasate from VEGF-loaded

meshes. Cells stained with calcein-AM. Scale bar is 200 µm. Graph

displayed quantifies the network formation over 5 days of VEGF release.

* indicates statistical differences with respect to the network formation

induced by blank releasate at each time point. ...............................................98

Figure 4.4: In vivo evaluation of VEGF release kinetics from electrospun gel-MA in

a rat subcutaneous model. A) Mass loss of blank and VEGF-loaded

gelatin-methacrylate meshes over 6 weeks. B) Corresponding release of

VEGF from gelatin-methacrylate meshes. .....................................................99

Figure 4.5: Schematic of co-electrospinning apparatus. Blow out image depicts dual-

fiber population. Fluorescein (green) fibers are gel-MA and DAPI (blue)

fibers are PLGA. ...........................................................................................100

Figure 4.6: Stress-strain response for each electrospun wrap under tensile loading.

Blue line indicates electrospun gel-MA, red line indicates electrospun

PLGA, and green line indicates co-electrospun gel-MA and PLGA.

Arrows denotes tensile failure at the associated strain. ................................101

Page 21: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

xxi

Figure 4.7: In vitro release of VEGF in PBS over 14 days. A) Cumulative release of

VEGF from electrospun gelatin-MA as compared to release from the co-

electrospun wrap. B) Corresponding daily release of VEGF from gel-MA

as compared to the co-electrospun wrap. Red-dashed line indicates the

lowest-targeted VEGF concentration. ..........................................................103

Figure 4.8: In vitro release of gentamicin sulfate in water over 14 days. A)

Cumulative release of gentamicin sulfate from electrospun PLGA as

compared to release from the co-electrospun wrap. B) Corresponding

daily release of gentamicin sulfate from PLGA as compared to the co-

electrospun wrap. Red-dashed line indicates the h ypothesized MIC. .........103

Figure 4.9: Fabrication of dopamine-modified PLGA wrap for tissue adhesion. A)

Schematic of the dopamine dip-coating process. B) ATR-FTIR of the un-

modified PLGA mesh (light gray line), pure dopamine (dark gray line),

and dopamine-modified PLGA mesh (black line). ......................................105

Figure 4.10: Evaluation of a dopamine-modified wrap on tissue adhesion. A)

Schematic of proposed reaction with the periosteum. B) Representative

image of the set-up for lap shear testing the dopamine-modified PLGA

mesh with bone. C) Average maximum shear strength of the dopamine-

modified PLGA mesh coated with increasing concentrations of

dopamine. Results of the un-modified PLGA mesh were not included in

the graph due to the shear forces being below the detection limit of the

instrument. * indicated statistical differences with respect to the 2 mg/mL

coating concentration. ..................................................................................106

Page 22: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

xxii

Figure A.1: Iterative design of a composite bilayer wrap with requisite mechanical

properties. A) Schematic of composite bilayer wrap fabrication for bulk

hydrogel-fiber composite, electrosprayed hydrogel-fiber composite, and

hydrogel foam-fiber composite. B) Set up for mechanical testing of the

three composite formulations and C) resulting stress-strain response for

each composite bilayer wrap (arrows indicate tensile failure at the

associated strain). *Figure created with BioRender.com. ............................140

Figure A.2: Characterization of the hydrogel foam + TMPETA- fiber composite. A)

ATR-FTIR of the gelatin layer (gray line) and the hydrogel layer (black

line). B) Cross-sectional SEM depicting the intra-microarchitecture of

each layer (scale bar =30 µm). SEM blowouts display the plan view of

each layer (scale bar =100 µm). C) Representative images of hDF

attachment over 14 days (scale bar =200 µm) and quantified cell adhesion

on each layer. Cells stained with rhodamine phalloidin (F-

actin/cytoplasm) and SYBR green (DNA/nucleus). * indicates statistical

differences with respect to the hydrogel foam at each time point, (two-

way ANOVA with Sidak’s analysis, P<0.05). .............................................142

Figure A.3: In vivo intra-abdominal adhesion study results. A) Schematic of rat in vivo

abrasion model utilized to assess adhesion formation. B) Composite

adhesion scoring for all treatments, red data points indicate observed

displacement of the treatment specimen. * indicates statistical differences

as compared to Interceed®, (P<0.05). Representative images taken from

site where treatments were applied: C) PBS treated, D) Tisseel® treated,

E) gelatin mesh treated, F) hydrogel foam treated, G) composite bilayer

treated, H) Interceed® treated. *Figure created with BioRender.com ..........146

Page 23: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

xxiii

Figure A.4: Effect of dopamine coating on tissue adhesion to the bilayer wrap. A)

Schematic of dopamine coating process and hypothesized reaction of

dopamine coating with the wound site. B) Schematic depicting the lap

shear test set-up and C) average maximum shear strength of the

dopamine coated bilayer wrap on porcine substrates as compared to

Interceed®. Un-modified gelatin control was not included in graph due to

shear forces below the detection limit of the instrument. * indicates

statistical differences as compared to Interceed®, (student’s t-test,

P<0.05). *Figure created with BioRender.com. ..........................................148

Figure A.5: Characterization of the dopamine-modified gelatin mesh. A) ATR-FTIR

of the un-modified gelatin (black line), pure dopamine (dark gray line),

and dopamine-modified gelatin mesh (light gray line). ...............................149

Page 24: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

1

Chapter I: A Critical Review of Biomaterial Approaches for Improved

Bone Regeneration with the Masquelet Technique

1.1 BONE LOSS MANAGEMENT

Traumatic injuries to the long bones account for approximately 6% of delayed-

union and non-union fractures, indicated by a period of bone bridging exceeding 6 months

and incomplete bone bridging, respectively [1, 2]. Non-union fractures not only impose a

medical burden on the patient but also an economic burden with an estimated

hospitalization cost over $30,000 per patient [1]. Bone salvaging procedures have been

implemented to reduce the rate of delayed unions and non-unions. One common bone

salvaging procedure is the Ilizarov bone transport technique [3]. This technique was first

introduced in the 1950s by Gavriil Ilizarov and uses distraction osteogenesis to fill defect

voids [4]. It entails sectioning the bone and using an external fixation device to gradually

separate the cut ends to allow new bone to bridge the gap [5, 6]. The Ilizarov technique has

a bridging rate of 83% to 100%; nonetheless, this technique has lengthy recovery times,

high rate of pin-site infection (≤ 80%), poor alignment, and poor bone consolidation [6].

Another common procedure is the vascularized fibular autograft technique, first

implemented in 1975 by Gian Taylor [7]. It involves microsurgical attachment of a free

vascularized fibula to the vasculature surrounding the defect [7]. This technique offers

immediate blood supply to the damaged tissue, over 30 cm of viable cortical bone,

immediate soft-tissue coverage, and reduced donor site morbidity [8]. Despite a bridging

Page 25: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

2

rate of 88% to 100%, this technique is limited by infection, stress fracture, and requisite

microsurgical expertise [6, 9]. An alternative approach to the aforementioned procedures

is the induced membrane technique. This technique was first described by Alain Masquelet

in 1986 and is now coined as the Masquelet technique. This two-stage procedure prompts

reconstruction of segmental bone defects utilizing a biological induced membrane,

morselized bone autograft, and bone marrow aspirate [3, 10]. In the first stage, a

polymethylmethacrylate (PMMA) cement spacer is implanted to prevent ingrowth of

fibrous tissue. Implantation of the PMMA spacer also stimulates a host response

characterized by cell infiltration and edema, which is then followed by acute inflammation,

chronic inflammation, and granulation tissue development that generates a fibrous capsule

(induced membrane) [11]. In the second stage, which takes place 6 to 10 weeks after the

first stage, the cement spacer is removed and is replaced with a mixture of morselized

cancellous autograft and bone aspirate harvested using the reamer-irrigator-aspirator

technique [2, 12]. The induced membrane that is formed during the second stage is used to

encapsulate the autograft during healing. The bridging rate for this procedure is comparable

to the Ilizarov technique and the vascularized free fibular autograft techniques (82 to

100%); however, the Masquelet technique does not impinge on daily activities, delay

weight bearing, or require technical expertise to the extinct of the other procedures [4, 9].

Despite the advantages over the other bone salvaging procedure, this technique is limited

by unpredictable clinical outcomes. Since its development, there have not been any

significant technical modifications to the Masquelet technique to address this limitation.

Page 26: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

3

The failure to address this limitation creates an opportunity for further advancement to

improve and standardize healing.

1.2 BIOLOGICAL ROLE OF THE INDUCED MEMBRANE

It has been suggested that the induced membrane has a significant role in healing

during the Masquelet technique [11, 12]. Therefore, researchers have focused on

elucidating the biological role of the induced membrane to provide the fundamentals for

further innovation and improve outcomes. Histological analysis indicated that the 1-2 mm

thick membrane consist of three distinct layers, primarily composed of fibrous extracellular

matrix, a vascular network, cells, and paracrine factors [9, 13-16]. These features provide

a favorable environment for bone regeneration similar to the native periosteum However,

the induced membrane is formed by a variable host response that results in inconsistent

membrane compositions, which have been found to contribute to the unpredictable clinical

outcomes [17]. This membrane variability has lead to researchers investigating alternative

methods to guide formation of the induced membrane.

1.2.1 Characterization of the Induced Membrane

The matrix of the induced membrane is primarily composed of collagen type 1 and

elastin fibers which are responsible for the high tensile strength, toughness, and elasticity.

Accumulation of these fibers over time allows for surgical handling, provides mechanical

stimuli to cells for mechanotransduction, and serves as a barrier to protect the autograft

from resorption [9]. The vasculature of the membrane provides transport of signaling

molecules, nutrients, and waste to support cells during bone remodeling. It also facilitates

Page 27: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

4

the transport of gases to help sustain the cell viability in the induced membrane [15, 18].

Blood vessels begin to form in the outermost layer, with vascularity seen as early as 2

weeks and peak density around 4 to 6 weeks after implantation [9]. The membrane is also

a source of bone marrow-derived stem cells measured by the presence of STRO-1-positive

cells [9, 15]. They are most prevalent in the outermost membrane and can be detected as

early as 2 weeks post-implantation [15, 19]. A subset of the cells in the membrane have

been shown to express markers of both embryonic and adult stems cells [20]. These cells

are capable of differentiating down osteogenic and chondrogenic lineages to further aid the

repair of injured tissue [15, 19]. Paracrine factors are also prevalent in the membrane. These

are soluble proteins (e.g. growth factors, cytokines) that are secreted by cells or transported

through the vasculature to induced cellular responses in nearby cells [15, 21, 22].

Angiogenic and osteogenic paracrine factors are detectable as early as 2 weeks, with levels

peaking between 4 and 6 weeks [15, 16, 21].

1.2.2 Limitations of the Induced Membrane

Unpredictable clinical outcomes are often due to the transient bioactivity and the

variable durability of the induced membrane. One of the most notable features that affects

the bioactivity of the induced membrane is the vasculature density. Approximately 40% of

the vasculature density decreases after 6 weeks [15, 16, 22, 23]. Vascular degeneration

causes a reduction nutrient, and waste transport which limits the healing capacity of the

membrane. A reduction in blood transport during the second stage of the procedure also

deprives blood-circulating paracrine factors to the transplanted MSCs and renders

Page 28: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

5

autologous bone graft at risk for necrosis [24]. Surgical approaches that have been

suggested to improve the vasculature require tradeoffs with soft tissue healing and

durability. For example, researchers have advocated for the second stage of the procedure

to occur between 4 to 6 weeks, as opposed to 6 to 10 weeks, to capitalize on the peak

bioactive potential of the induced membrane. However, this tradeoff would compromise

the time required for soft tissue healing. Clinicians have postulated that epithelization and

revascularization of the soft tissue surrounding the defect occurs during the 6 to 10-week

period [25-28]. This time is also necessary to establish the mechanical properties of the

membrane, which are primarily determined by the composition of the extracellular matrix

secreted by adherent inflammatory cells (i.e. macrophages and fibroblasts) during the host

response [11, 15, 25]. As the degree of the host response can vary over this period,

inconsistency in durability occurs across cases [29]. The anatomical location of the defect

also has a role in the variability of the vasculature and the durability of the induced

membrane [25, 29, 30]. These variances can adversely impact surgical handling, barrier

properties, and mechanotransduction. Furthermore, the harmful effects of microbial

infection present another limitation that impacts the bioactive potential of the induced

membrane. Inadequate debridement during the first stage often leads to persistent infection.

This can cause chronic inflammation and tissue necrosis which impedes reconstruction and

requires a revision surgery. Overall, the degree of these limitations can vary across patient

populations which makes achieving predictable clinical outcomes challenging [6, 9, 26].

Page 29: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

6

1.2.3 Recent Approaches to Guide Membrane Formation

Currently, no commercial products exist to standardize formation of the induced

membrane for improved bioactivity and durability. Researchers have attempted to modify

the properties of the induced membrane or inhibit infection for improved clinical outcomes

via alterations to the PMMA spacer [31-36]. Modification of PMMA spacer topography

was evaluated in an aim to enhance vascularization via an increase in membrane surface

area. This approach successfully increased the surface area of the induced membrane but

no assessment was performed to validate the effect on vascularization. There was also no

significant difference in bone formation as compared to treatment with a PMMA spacer

[34]. In another study, a calcium sulfate spacer was investigated as an alternative solution

to improve expression of growth factors that regulate angiogenesis and osteogenesis. This

approach was unsuccessful at enhancing expression and did not improve bone regeneration

as compared to a PMMA spacer [31]. Others have similarly evaluated the effect of spacer

material on membrane formation [32, 33, 35]. A titanium spacer generated an induced

membrane with biochemical expression comparable to a PMMA-induced membrane.

However, this membrane did not promote autograft integration as well as the PMMA-

induced membrane [33]. The titanium spacer was later evaluated with roughened

topography as a means to improve durability and biochemical expression of the membrane

[32, 35]. The roughened titanium space produced a more durable membrane than a PMMA

spacer with a 40% increase in tensile strain by 40% and a 58% reduction in the elastic

modulus without changing tensile strength or toughness. These results were attributed to

the isotropic mechanical properties of the membrane under tensile stress and indicated that

Page 30: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

7

roughened topography improved the durability of the induced membrane, such that the

membrane can deform during surgical handling while retaining integrity. Careful

consideration should be given to the use of roughened spacers due to the significant

difference between the mechanical properties of the resulting membrane and the native

periosteum [32, 37]. The anisotropic mechanical properties of the periosteum exerts

mechanical stimuli to progenitor cells involved in osteogenesis that may not be present

with the more durable induced membrane [37]. A follow-up study confirmed that the

durable membrane did not improve biochemical expression and was inferior to the

performance of a thinner membrane induced by a PMMA spacer in bone regeneration [35].

Furthermore, antibiotic-loaded PMMA spacers were investigated and exhibited infection

clearance sufficient to restore biochemical expression comparable to non-infected induced

membranes. Nevertheless, the antibiotics did not enhance expression as to improve

treatment over the standard technique [36]. These failed attempts to enhance angiogenesis

and biochemical expression in the induced membrane highlight the need for a method to

better guide formation of the induced membrane during the Masquelet technique. An ideal

approach would guide membrane formation with a focus on the following key design

criteria: improve durability, enhance vascularization, and provide infection control.

1.3 IMPROVE MECHANICAL DURABILITY

Variations in the durability of the induced membrane significantly contribute to the

limitations of the Masquelet technique. It is necessary to standardize the durability to

improve surgical handling, protect the autograft from resorption, and provide proper

Page 31: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

8

mechanical stimuli to progenitor cells. As previously described, the mechanical durability

of the induced membrane is controlled by the matrix secreted by macrophages and

fibroblasts during the host response to the spacer [11, 15, 25]. An adjunct substrate that can

provide a framework for cellular attachment and a template for guiding matrix deposition

and tissue remodeling has the potential to improve durability. One of the most notable

tissue engineering approaches used to guide cellular interactions for remodeling is

biomaterial scaffolding. Biomaterial scaffolds are often designed to mimic the fibrous and

porous microarchitecture of the matrix. The high surface-area-to-volume ratio of the

fibrous constituents selectively enhance adsorption of additional serum proteins that

promote cell attachment [38-40]. In addition to the ability of the microarchitecture to

enhance cellular attachment, the material properties of fibrous scaffolds can provide

biochemical cues to further guide cellular behavior during remodeling [39]. There are three

main fabrication techniques used to generate fibrous scaffolds which consist of freeze-

drying, microfluidic spinning, and electrospinning. Each of these techniques offer tunable

scaffold properties through material selection and processing conditions that can be used

to direct formation of the induced membrane with improved durability.

1.3.1 Freeze-drying

Freeze-drying is a form of thermally-induced phase separation. It involves freezing

a polymer solution at temperature below the freezing point of the solvent. This causes the

polymer to coalesce leading to a polymer-rich and polymer-free phase. The frozen solution

is then subjected to sublimation in which the solvent transitions directly from a solid state

Page 32: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

9

to a gas state under a low pressure leaving a porous, fibrous polymer-rich network. Fiber

diameters of freeze-dried scaffolds range from nanometers to microns [38, 41, 42]. The

fiber diameter can be tuned by modification to the polymer concentration and the freezing

temperature. Lower polymer concentrations result in smaller fiber diameters due to a lower

polymer-rich phase. Similarly, freezing at lower temperatures results in smaller fibrous

structures [42]. Freeze-dried scaffolds are typically fabricated using water-soluble natural

and synthetic polymers. This is an advantage for applications that require cellular

interactions, as natural polymers impart bioactive sites that promote cell adhesion and

guide cellular behavior. Additionally, these polymers are resorbable which enable

remodeling [43]. The mechanical properties of the scaffolds can be strengthen through

crosslinking before or after freeze-drying [44]. Crosslinking can also be used to control the

resorption rate during remodeling. Despite their tunable fibrous properties, freeze-drying

can result in a laminar sheet-like microarchitecture instead of fibers if the polymer

concentration and freezing are not well controlled [42].

1.3.2 Microfluidic Spinning

Microfluidic spinning is a common manufacturing process involving an aqueous

polymer solution that flows through an oil-based sheath or in a silicone microchannel [45-

48]. Differences in flow rates, surface tension, and energy dissipation keeps the two

streams separated. This technique allows for precise control over the architecture and

uniform size of the resultant fibers. Microfluidic spinning produces fiber diameters that

range from nanometers to hundreds of microns, comparable in diameter to fibrils of the

Page 33: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

10

native matrix [45]. Similar to freeze-drying, material selection for microfluidic spinning is

often limited to water-soluble synthetic and natural polymers. The mechanical properties

of these scaffolds are governed by crosslinking performed after precipitation in a

coagulation bath or in situ during flow [45]. Further tuning of mechanical properties can

be achieved by using a rotating spool in the coagulation bath. A rotating spool collects

fibers along a unidirectional axis leading to aligned fibrous matrices. Fiber alignment along

the axial direction of the loading force often results in higher tensile properties due to

greater resistance of fiber reorientation. Fiber alignment can modulate cell attachment, and

thus, can control directionality of the matrix to further control mechanical properties [49].

A rotating spool can also be used to control mechanical properties through modulation of

the fiber diameters. Higher rotational rates often lead to smaller fibers with greater

mechanical strength due to less architectural defects, as compared to larger fibers generated

at lower rates [50]. Smaller fibers can also permit greater cell infiltration due to larger pores

created by reduced fiber packing density [51]. Although microfluidic spinning offers many

advantages for cell adhesion and guiding cellular behavior, the hydrogel-like properties

limits its use in applications requiring structural support [45].

1.3.3 Electrospinning

The most common fabrication technique for producing fibrous scaffolds is

electrospinning, during which, an electric potential is applied to a polymer solution that is

constantly flowing from a syringe. Charge repulsion within the solution droplet at the end

of the capillary overcomes the solution surface tension leading to a polymer jet erupting

Page 34: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

11

from the droplet towards a ground or oppositely charged collector [52]. The solution

parameters and electrospinning set-up can be modulated to generate fiber diameters that

range from nanometer to micron-sized fibers [45, 53].

The mechanical properties of electrospun fibers can be managed by the selection of

polymer, fiber orientation, collection time, and post-processing conditions. As previously

mentioned, natural polymers are chosen over synthetic polymers for constructs that require

greater bioactive sites to guide cell behavior [45, 54]. Furthermore, the fiber orientation

can be configured based on the conditions of the collector to control cellular alignment

[54]. Rotating mandrels at relatively high speeds often result in aligned electrospun meshes

with greater mechanical properties than electrospun meshes collected on a static plate [54].

An inherent limitation of electrospinning is the high fiber packing density that limits cell

infiltration due to small pores [54]. However, this can be overcome by using various

electrospinning set-ups such as co-electrospinning and co-axial electrospinning, which

enable the combination of materials to harness multiple material properties in a single

construct [54]. Materials with varying resorption rates can be combine such that a faster

resorption rate will reduce the fiber packing density and enable cell infiltration [51].

Although the electrospinning set up is highly versatile, sensitivity to ambient conditions

requires frequent modifications to the electrospinning set up [45].

1.3.4 Fibrous Scaffolds to Improve Durability

In summary, improving the durability of the induced membrane requires a

resorbable substrate that provides a template to guide cellular attachment and matrix

Page 35: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

12

deposition as well as direct cellular behavior during tissue remodeling. Fibrous scaffolds

are ideal candidates due to their structural similarity to the native matrix which can drive

cellular interactions. The most commonly used fabrication techniques to generate fibrous

scaffolds include freeze-drying, microfluidic spinning, and electrospinning. These

techniques each offer unique and tunable properties such as biochemical signaling, fiber

diameter, and mechanical strength that can be used to improve durability of the induced

membrane.

1.4 ENHANCE VASCULARIZATION

In addition to increasing the durability of the induced membrane, it is also

imperative to enhance the vasculature to increase the bioactive potential of the membrane

during the second stage. Formation of the vasculature is primarily controlled by

angiogenesis which is a process where new vessels form from neighboring vessels [24].

This process is tightly regulated via the Notch-1 pathway which controls proliferation and

differentiation of endothelial cells. However, it is suggested that this pathway becomes

unregulated during the formation of the induced membrane leading to vessel degeneration

[22]. The transport of angiogenic factors decreases as a result of vasculature degeneration,

which impedes vascularization during the second stage of the procedure and delays bone

regeneration [15, 16, 21, 22]. Therefore, incorporation of bioactive cues into the adjunct

fibrous substrate to enhance angiogenesis will be important to increase the vasculature.

Recent approaches to enhance angiogenesis have focused on delivery of angiogenic

Page 36: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

13

factors, gene delivery, integrin-targeting, and cell delivery, with many of these approaches

overlapping.

1.4.1 Delivery of Angiogenic Factors

Controlled release of angiogenic factors from polymeric matrices has been

investigated as a therapy to improve angiogenesis. This approach circumvents the adverse

effects of short growth factor half-lives, growth factor dilution in blood plasma, and

systemic toxicity associated with high levels of growth factor [55, 56]. The desired release

kinetics of the angiogenic factor from the polymer matrix helps to guide selection of the

polymer type and the technique for matrix fabrication [57].

Synthetic matrices offer several advantages for angiogenic factor delivery

including ease of fabrication, tunable degradation, and established use in controlled

delivery [58]. However, the harsh processing conditions required for fabrication of these

matrices, such as high temperatures or organic solvents, can denature factors leading to a

loss in bioactivity [59]. To circumvent this loss of bioactivity due to processing, angiogenic

factors can be incorporated after fabrication by adsorption onto the surface or absorption

into the polymer matrix, with subsequent delivery governed solely by diffusion.

Nevertheless, post-fabrication loading can restrict the encapsulation efficiency thereby

reducing the potential efficacy of the treatment [60]. Another concern with the use of

synthetic matrices for controlled delivery is that degradation of them can result in an

inflammatory response due to toxic byproducts or changes to the local pH [6].

Page 37: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

14

As such, natural matrices and their derivatives are preferred over synthetic matrices

as they also offer ease of manufacture, tunable degradation, and established used in

controlled delivery. However, contrary to synthetic matrices, they are often processed in

aqueous solvents allowing for in-line loading of the angiogenic factors with a corollary

increase in encapsulation efficiency. Another advantage over synthetic matrices is that

degradation byproducts of biological materials are cytocompatible and are readily cleared

from the body [61, 62]. Nevertheless, controlled delivery from natural matrices is primarily

governed by an increase in the crosslink density or conjugation of the factor, resulting in

structural changes to the matrices or conformational changes to the factor, respectively

[63]. Affinity sequestration mechanisms have been explored as a means to sequester factors

for sustained release with minimal effect on the structural properties of natural matrices

and without loss of bioactivity. These mechanisms include extracellular matrix-derived

ligands (e.g. heparin, collagen-binding domains) [64, 65], aptamers [66], and nanomaterial

additives (e.g. nanodiamonds, nanoclays) [67, 68]. Despite the potential to sequester

angiogenic factors with minimal impact on the bioactivity, careful consideration must also

be given to the transient and reversible interactions that govern sequestration when

sustained preservation is desired [69]. Furthermore, it is difficult to mimic the endogenous

regulation of protein expression with delivery of angiogenic factors from biomaterials

carriers [70].

Table 1.1: Summary of angiogenic factors indicated to regulate angiogenesis.

Angiogenic factor Role in angiogenesis References

Page 38: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

15

TNF-α

Cytokine family;

Activates inflammatory phase;

Upregulates expression of angiogenic factors in

inflammatory cells

[71, 72]

VEGF

Growth factor;

Stimulates proliferation and migration of

endothelial cells;

Stimulates formation of capillary like structures

[73, 74]

FGF-2

Growth factor;

Stimulates proliferation and migration of

endothelial cells;

Recruits pericytes;

Promotes matrix depositions for blood vessels;

Upregulates expression of VEGF;

[73, 74]

TGFβ-1

Cytokine;

Activates inflammatory phase;

Increases expression of angiogenic factors in

inflammatory cells

[71, 74]

PDGF Upregulates expression of VEGF [73, 74]

ANG 1/2

Promotes vessel maturation;

Mediates migration, adhesion and survival of

endothelial cells;

Disrupts the connections between the endothelium

and perivascular cells;

Promotes cell death and vascular regression;

Promotes neovascularization in the presence of

VEGF

[74, 75]

Page 39: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

16

Common angiogenic factors used to enhance angiogenesis include cytokines and

angiogenic growth factors such as tumor necrosis factor-alpha (TNF-α), vascular

endothelial growth factor (VEGF), fibroblast growth factor (FGF), transforming growth

factor beta-1 (TGFβ-1), transforming platelet-derived growth factors (PDGF), and

angiopoietin (ANG). A subset of these factors have a direct role in initiating angiogenesis

(i.e. VEGF, FGF, TGFβ-1) whereas others regulate expression of angiogenic factors (i.e.

ANG, PDGF, TGFβ-1, TNF-α) (Table 1.1) [72, 74].

1.4.2 Gene Delivery

The limitations associated with delivery of angiogenic factors (i.e. timing of release

and dosing) introduce challenges for improving angiogenesis in tissue engineering

applications. Recent innovative strategies encompassing genetic engineering offer an

alternative solution to the delivery of angiogenic factors [70]. Specifically, gene delivery

enables foreign genetic material encoded for angiogenic factors to be integrated into the

host genome or replicated independently of it to induce overexpression of the gene. This

technique enables endogenously sustain levels of the selected angiogenic factor for

enhanced angiogenesis. There are two primary methods of delivery for genetic material: 1)

viral vectors and 2) non-viral vectors [76].

Viral vectors are regarded as those that integrate with the host genome.

Adenoviruses are the most clinically used viral vectors for gene delivery. They are non-

enveloped viruses containing dsDNA [76, 77]. The ability of adenoviruses to integrate with

the host genome enables high transfection rates and sustained expression of the angiogenic

Page 40: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

17

factor. However, integration into the host genome increases the risk of provoking an

immune response. Non-viral vectors are those that do not integrate with the host genome.

The most commonly used non-viral vectors are plasmids which are bacterial dsDNA

molecules [77]. Plasmid vectors are regarded as the safest carriers as they do not integrate

with the host genome and have rapid clearance from the body. Although plasmid vectors

are safer than adenoviruses, their bioactive potential is limited by their transient presence

in the body [76, 77]. The risk of inducing an immune response and the limited transfection

efficiency have shifted the focus gene delivery to biomaterial matrices.

Biomaterial matrices offer protection and tunable delivery of genetic material to

host cells. Polymeric matrices are especially beneficial for entrapment and sustained

delivery of sensitive genetic material, as versatility in material selection and corollary

processing conditions broaden the mechanisms governing sequestration and release, as

previously described. As compared to conventional carriers, polymeric matrices have been

demonstrated to enhance expression of angiogenic factor and angiogenesis in tissue

ischemia [78]. Common polymeric matrices that have been investigated include hydrogels

[79-81], nanoparticles [82, 83], and porous constructs [84]. Despite the demonstrated

potential of polymeric matrices, degradation byproducts can induce an immune response,

as previously described [85].

1.4.3 Integrin Targeting

The adhesive interactions of vascular endothelial cells with the matrix aids in

regulation of angiogenesis [86, 87]. These interactions are governed by integrins, cell

Page 41: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

18

surface receptors composed of α and β subunits [87]. The specific mechanisms by which

integrins regulate angiogenesis is not well-understood; however, there is upregulation of

α5β1, αvβ3, α1β1, and α2β1 expression on endothelial cells during initiation of angiogenesis

[86, 88, 89]. Furthermore, synergistic interactions between growth factor receptors and

integrins have been demonstrated to improve angiogenesis during wound healing.

Inhibition of VEGF binding following integrin blocking confirmed that integrin-binding is

important in regulation of growth factor-induced angiogenesis [90]. These findings have

given rise to biomaterial approaches that combine integrin targeting with growth factor

delivery by incorporating ligands into biomaterial carriers for growth factors. Ligands are

molecules that form complexes with integrins to promote cellular responses. The

combination of ligand priming with growth factor delivery offers an innovative method

that encompasses biomimicry of this integrin-receptor crosstalk to improve angiogenesis

[86, 89].

Table 1.2: Summary of ligands commonly used for integrin targeting of vascular

endothelial cells.

Peptide/protein ligand Targeted integrin References

RGD;

Fibronectin;

Vitronectin;

Fibrinogen

α5β1;

αvβ3 [88, 89]

GFOGER;

Collagen-1

Laminin

α1β1;

α2β1;

[86, 89]

Page 42: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

19

Selection of ligands with high specificity towards the aforementioned integrins is

critical for fabrication of effective angiogenic biomaterials [49]. Ligand priming for

production of angiogenic biomaterials is typically achieved by incorporation of peptides or

proteins summarized in Table 1.2 [87, 89]. These ligands have been incorporated into

biomaterial matrices using chemistries such as carbodiimides [91], periodate oxidation

[92], and Diels-Alder chemistries [93] and have been demonstrated to improve

vascularization [89]. Although ligand priming has the potential to improve angiogenesis

by imparting bioactivity into biomaterial matrices to mimic the interactions between cells,

matrix, and growth factors, there is concern with negative regulation of angiogenesis due

to peptide specificity [94].

1.4.4 Cell Delivery

Vascular endothelial cells and endothelial progenitor cells have vital roles in

angiogenesis. Their ability to organize into new vascular networks or integrate into existing

networks places them at the forefront of tissue engineering strategies for angiogenesis.

Researchers have turned to biomaterial scaffolds that enable sequestration of cells due to

the transient retention that results from direct transplantation [70]. Hydrogel-based

scaffolds are excellent candidates for angiogenic cell delivery as their mild processing

conditions enable cell encapsulation. They can be fabricated into architectural templates

that encourage cell organization into new vasculature networks [95, 96]. Furthermore,

modulation of the chemical and physical properties of hydrogels enables tunable release

kinetics for applications requiring cell release and integration into existing vasculature

Page 43: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

20

networks [97]. Non-hydrogel-based scaffolds also provide platforms for which seeded cells

can proliferate, migrate, and organize into new vasculature networks. Similarly, material

properties, such as the mechanical strength, can be altered to influence cell behavior and

organization [98]. Although cell encapsulation and structural support are critical design

requirements for transplantation, they are only effective if cell viability is maintained [70].

A key factor responsible for failed cell delivery approaches is hypoxia, which is a

condition described by low-oxygenated environments. This is particularly true for cell-

encapsulated scaffolds that have low blood perfusion [70, 99]. Several groups have

investigated methods to enable oxygen production within the constructs. These methods

include embedment of inorganic peroxide species that interact with water to form hydrogen

peroxide and oxygen as an intermediary products [100-102]. Incorporation of inorganic

peroxide species has been shown to increase cell viability, with sustained oxygen level for

up to 10 days [101]. However, high concentrations of hydrogen peroxide pose a safety risk

for these approaches [99].

1.4.5 Mechanisms to Enhance Vascularization

Overall, increasing the vasculature is required to improve the bioactive potential of

the induced membrane at later stages of the Masquelet technique. Researchers have

developed several mechanisms to impart bioactive cues to increase the vasculature through

angiogenesis. These mechanisms include delivering angiogenic factors from polymeric

carriers, inducing expression of angiogenic factors via gene delivery from viral and non-

viral vectors, guiding endothelial cell behavior via integrin targeting biomaterials, and

Page 44: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

21

delivering angiogenic cell lines using polymeric carriers. In some cases, these techniques

can be combined to generate a synergistic effect in angiogenesis. Incorporating these

mechanisms into a fibrous scaffold can generate a substrate that not only improves the

durability but also enhances angiogenesis in the induced membrane for sustained

bioactivity during treatment.

1.5 PREVENT OSTEOMYELITIS

Although guiding formation of the induced membrane to enhance the durability and

the vasculature will improve the Masquelet technique, preventing osteomyelitis remains a

challenge. Infection recurrence is one of the leading complications of the Masquelet

technique [6, 103, 104]. Despite radical debridement, up to 30% of reconstruction failures

are attributed to infection recurrence. Inadequate debridement is often followed up with a

second pass of debridement accompanied by systemic delivery of antimicrobials [6, 104,

105]. However, moderate levels of infection clearance result due to dilution of the

antimicrobials in the blood, off-target tissue absorption, and poor blood circulation around

the bone defect [106, 107]. High and potentially cytotoxic doses of antibiotics are

administered to overcome the potential loss due to systemic delivery. The reduced efficacy

and risk of toxicity has led clinicians to explore local antimicrobial delivery as an

alternative solution. Local delivery of antimicrobials can more precisely target the infected

tissue at greater concentrations that would normally be reduced via systemic delivery,

while avoiding systemic toxicity [107]. Furthermore, local delivery can expand the

selection of antimicrobials available for treatment of osteomyelitis.

Page 45: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

22

1.5.1 Antibiotics

Since its discovery, antibiotics have become the standard choice among

antimicrobial agents used for infection control. They have been broadly used since

penicillin was introduced during the 20th century, and have been proven to effectively kill

bacteria primarily by preventing bacterial cell wall formation and inhibiting protein

biosynthesis and DNA replication [108-110]. Selection of antibiotics largely depends on

its bacterial spectrum activity, antibiotic sensitivity, and the route of administration [111,

112]. Among its diverse classes, the most commonly administered antibiotics are beta-

lactam antibiotics, glycopeptides, aminoglycosides, tetracyclines, fluoroquinolones, and

rifampicin [112]. Their corresponding mechanisms of action are summarized in Table 1.3.

Table 1.3: Summary of the common classes of antibiotics indicated to treat bone

infection.

Antibiotic

Class

Common

Forms

Mechanism

of Action

Spectrum

Activity References

Aminoglycosides

Gentamicin,

amikacin,

streptomycin

Inhibits protein

synthesis by binding

irreversibly to

bacteria’s 30S-

ribosomal subunit

Creates fissures in

bacterial cell

membrane causing

cell leakage and

increased antibiotic

uptake

Gram-

negative

bacteria

[112-114]

Page 46: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

23

Beta-lactams

Penicillin,

oxacillin,

cephalosporin

Prevents bacterial

cell wall formation

by binding to the

active site of the

transpeptidase

Gram-positive

bacteria

[112, 115,

116]

Fluoroquinolones

Levofloxacin,

ciprofloxacin,

ofloxacin

Inhibits DNA

synthesis and

replication by

inhibiting DNA

topoisomerases

Gram-positive

bacteria [112, 117]

Glycopeptides Vancomycin

Prevents bacterial

cell wall formation

by inhibiting

transpeptidase and

peptidoglycan

synthesis

Gram-positive

bacteria [112, 118]

Rifampicin Rifampin

Suppresses

initiation of RNA

synthesis by

inhibiting DNA-

dependent RNA

polymerase activity

Mycobacteria [112, 119]

Tetracyclines Tetracycline,

Minocycline

Inhibits protein

synthesis by

preventing

aminoacyl-tRNA

from binding to the

ribosomal receptor

Gram-positive

and gram-

negative

bacteria

[112, 120]

Clinicians have routinely administered antibiotics to treat bone infection [108, 121].

The efficacy of gentamicin, cephalosporin, and levofloxacin have been demonstrated

Page 47: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

24

against Staphylococcus aureus, including methicillin-resistant strains commonly known to

cause bone infection [122-125]. Similarly, experimental models of bone infection have

been successfully treated using vancomycin alone or in combination with rifampin [126,

127]. The established utility and proven effectiveness of antibiotics inarguably place them

at the forefront of antimicrobial agent selection [59, 123, 127, 128]. The long history of

antibiotics also provides extensive evidence that offers highly specific and supported

mechanisms of action against bacterial pathogens [109, 110, 121]. However, its widespread

administration throughout the decades raises concerns for bacterial resistance [108, 129] .

1.5.2 Metals

The therapeutic use of metal compounds dates back to ancient civilization.

However, the discovery and widespread use of antibiotics halted advancements in

developing metallic antimicrobial agents [130, 131]. With the rising incidence of antibiotic

resistance, there is resurgence in scientific interest of metals for infection prevention of

infection [131, 132]. Recent findings have proposed that metallic antimicrobials eliminate

pathogens by acting on a combination of metabolic processes such as replication,

transcription, translation. They inhibit these processes by producing reactive oxygen

species, disrupting the cell wall, protein dysfunction, and interfering with iron-dependent

pathways [130, 131, 133]. Various types of metals have been explored and include cationic

forms of silver, zinc, and gallium [134]. Their corresponding modes of action are

summarized in Table 1.4.

Table 1.4: Summary of metallic antimicrobials indicated to prevent bone infection.

Page 48: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

25

Metal Common Applications Mechanism of Action References

Silver

Topical burn treatment,

wound dressings,

antimicrobial coating for

implants and orthopedic

fixtures

Produces reactive oxygen

species that induces bacterial

cell death;

Reacts with peptidoglycans

that puncture and destroy

bacterial cell wall;

[130, 133,

135]

Zinc

Topical treatment for

dermatologic conditions

(e.g. acne vulgaris,

dermal infection,

dandruff), dental

applications, oral rinses,

and nanoparticles

Generates reactive oxygen

species that leads to bacterial

cell death;

Reduces ATP synthesis and

inhibits enzymes critical to

cellular activity;

[136, 137]

Gallium Topical ointment for

burns, wound dressings

Inhibits bacterial and fungal

growth by interfering with

iron-dependent processes;

Competitively inhibits

binding of Fe (III) and

deprives the target pathogen

of this essential nutrient

[138-141]

The clinical use of metallic antimicrobials is limited; however, current research

presents its potency against a wide range of bacterial and fungal pathogens that cause bone

infection [136, 139]. In experimental models of osteomyelitis and periprosthetic infection,

silver and zinc have demonstrated efficacy against a broad spectrum of microorganisms

including both gram-negative and gram-positive bacteria [142, 143]. Similarly, gallium has

been reported to reduce bacterial activity of Staphylococcus aureus when metallurgically

Page 49: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

26

added to titanium alloys, one of the most commonly used material for bone fixation devices

[144, 145]. This finding confirms that metallic antimicrobials are promising therapeutic

candidates for controlling orthopedic-related infections. Their broad-spectrum activity and

ability to exploit mechanisms independent of microbial metabolic pathways can potentially

address the ongoing challenge of antibiotic resistance. However, their exact mechanisms

of action and cytotoxic effects are not yet fully understood and effective dose

concentrations remain unclear [138, 142, 143].

1.5.3 Antimicrobial Peptides

In recent years, antimicrobial peptides (AMPs) have become viable alternatives to

conventional antibiotic treatment used in infection control [146]. AMPs are relatively small

molecules made of short sequences of amino acids that fold into an amphiphilic

configuration and behave as cationic species [146-148]. They contain approximately less

than 50 amino acids with almost 50% of the chain being hydrophobic species [146, 148].

Their overall net positive charge can be attributed to excess lysine and arginine residues,

and allows for preferential targeting of anionic bacterial cell membranes that causes pore

formation and membrane disintegration [147-149]. Despite similarities in general physical

properties, AMPs greatly vary in sequence and can be classified according to four main

categories of secondary structures whose structural characteristics and suggested modes of

action are summarized in Table 1.5 [150].

Table 1.5: Proposed structure-activity relationship of various antimicrobial peptides.

Page 50: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

27

Class Structural

Characteristics

Mechanisms of

Antimicrobial

Activity

Examples References

α-helical

Peptides

Unstructured in

aqueous

environment but

can form barrel-

like bundles in

bacterial membrane

Creates clusters of

AMPs or toroidal

pores on the bacterial

cell membrane that

disrupts its processes

Magainin,

Cecropin,

Pexiganan

[151-153]

β-sheet

Peptides

Rigid structures

stabilized by

disulfide bonds

Perpendicularly

penetrates and

destroys bacterial cell

membrane by

inducing pores

β -defensins,

Protegrins,

Tachyplesin

[150-152]

Extended

Peptides

Possess irregular

secondary

structures but

contains a high

percentage of

proline, tryptophan,

arginine, and

histidine amino

acid residues

Non-membrane

active; Penetrates

bacterial cell

membrane and

interacts with

intracellular proteins;

induces cell

membrane leakage

Indolicin [151, 152,

154]

Loop

Peptides

Loop formation

using a single

disulfide bridge

Suggested to use

stereospecific targets

but remains unclear

Thanatin [151, 155]

Studies that have explored the antimicrobial activity of AMPs against bone

infections have demonstrated efficacy against a broad spectrum of pathogens, including

antibiotic resistant strains like Staphylococcus aureus, Pseudomonas aeruginosa, and

Staphylococcus epidermidis [156-159]. Considering its broad-spectrum activity and

Page 51: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

28

selectivity in targeting pathogens, AMPs could potentially be adapted clinically as

antimicrobial agents [160]. However, AMPs currently remain comparably inferior to

conventional antibiotic treatment since their exact mechanisms of action are not firmly

established and concerns about acquired AMP resistance has been raised [148, 151, 155].

1.5.4 Resorbable Matrices for Antimicrobial Delivery

Local delivery systems for treatment of infection in orthopedic applications were

first introduced in bone cement stabilizers as a means to prevent infection during joint

arthroplasty procedures [161]. Bone cement has since become a long-standing approach to

implement local delivery of antibiotics, making its way into the Masquelet technique as an

antibiotic-embedded PMMA spacer [9, 28, 104, 162]. However, spacers fabricated from

PMMA bone cement are not resorbable, leading to poor release kinetics and the need for a

secondary removal procedure [36, 163, 164]. Another limitation with its use is that few

antimicrobials are able to withstand the exothermic conditions generated during

polymerization [36]. This limitation has peaked interested in the use of resorbable matrices.

Resorbable matrices used for local antimicrobial delivery are generally composed of

bioceramics or polymers. These materials have variable resorption rates, which prevent a

secondary removal procedure, and offer a method to tune release [107, 165]. Furthermore,

modifications to the physical properties such as surface-area-to-volume ratio, diffusion

path length, and swelling serve as additional methods to control release from resorbable

matrices [165].

Page 52: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

29

Bioceramics are a class of bone substitutes that are commonly used as bone-void

fillers. One common bioceramic that is used as an off-label carrier for antimicrobials in the

treatment of bone infection is calcium sulfate. As a bone substitute, osteoclasts readily

adhere to and infiltrate calcium sulfate resulting in bulk erosion. Bulk erosion allows

greater penetration of water which facilitates rapid release via diffusion within 24 h [165,

166]. An alternative bioceramic that is used off-label for antimicrobial delivery is calcium

phosphate. Resorption of calcium phosphate is relatively slow, as compared to calcium

sulfate, resulting in diffusion independent of the resorption rate. Release from calcium

phosphate is primarily influenced by the diffusion path length through the porous matrix

and spans over days to weeks [163, 165]. In general, diffusion from bioceramic matrices is

relatively quick and should only be considered for treatment of acute infection.

In contrast to bioceramic matrices, polymeric matrices offer sustained release of

antimicrobials for treatment of chronic infection via selection of polymer type (e.g.

synthetic and natural), as previously described. This is particularly evident in synthetic

matrices that primarily release antimicrobials via degradation-mediated diffusion.

Modulation of the polymer formulation can significantly alter the degradation rate leading

to diffusion on the order of weeks to months [107, 163]. If greater sustained release is

desired, then surface-eroding synthetic matrices are advantageous due to the zero-order

release kinetics that result [163]. Contrarily, release from natural matrices is less controlled

due to release being primarily governed by swelling-based diffusion [107, 163, 165]. This

mechanism of diffusion limits sequestration to antimicrobials that are smaller than the

swollen network mesh size; however, modifications to the crosslink density can reduce the

Page 53: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

30

mesh size and corollary swelling to provide better control over sequestration and release,

as previously described [167]. Nevertheless, one concern with the use of bioceramic and

polymeric matrices for antimicrobial delivery is that cytotoxic byproducts may exacerbate

the inflammatory response resulting in reduced efficacy of the antimicrobial [163, 165].

1.5.5 Local Delivery of Antimicrobials

In all, local delivery of antimicrobial can overcome limitations of osteomyelitis

associated with the Masquelet technique. Several different antimicrobials have been

demonstrated to be effective towards osteomyelitis. These include antibiotics, metal

compounds, and antimicrobial peptides. Controlled release of these antimicrobials is a key

design criteria for local treatment of infection. Researchers have investigated various

resorbable biomaterials such as bioceramics and polymers as carriers of antimicrobials and

have shown that these materials can be tuned to control release. As such, implementing

controlled delivery of an antimicrobial into the resorbable fibrous substrate for guiding

membrane formation can ultimately advance the Masquelet technique.

1.6 SUMMARY AND APPROACH

Despite high union rates with implementation of the Masquelet technique, there are

still challenges that need to be addressed for further advancement. Most notably, the non-

guided formation of the induced membrane yields variable membrane compositions with

unpredictable clinical outcomes. Of note is the mechanical durability of the induced

membrane. Although the membrane serves as a barrier to protect the autograft from

resorption, its mechanical properties vary across patient population which raises concerns

Page 54: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

31

with surgical handling and mechanotransduction. The combination of biomaterial matrix

design and biomimicry presents an opportunity to reduce structural variability of the

induced membrane and improve tissue remodeling. Additionally, enhancement of

angiogenesis during the second stage of the procedure continues to be the focus of research

for increased vascularization and improved bioactivity. New approaches developed to

address this limitation include delivery of angiogenic factors, gene delivery, integrin-

targeting ligands, and cell delivery. Furthermore, local delivery of antimicrobials with

tunable release kinetics will be necessary to prevent infection recurrence associated with

inadequate debridement. Several attempts have been made to guide formation of the

induced membrane with most consisting of modification of the spacer material and

topography. Despite these attempts, researchers have yet to discover a substrate that

concurrently addresses these limitations with successful improvement of clinical outcomes.

To this end, we propose to develop a multifunctional electrospun wrap composed

of poly(lactide-co-glycolide) (PLGA) and in situ-crosslinked gelatin that can concurrently

address the limitations of durability, vascularity, and infection associated with the

Masquelet technique. This adjunct wrap will provide a resorbable fibrous framework that

permits cell attachment and serves as template to direct matrix deposition and remodeling.

It also offers independent control over the release of multiple factors to simultaneously

prevent infection and promote angiogenesis. The following chapters describe our work on

developing this novel adjunct wrap. First, the limitations of conventional PMMA bone

cement used for local delivery prompted a comparative analysis of gentamicin sulfate and

gallium maltolate released from electrospun PLGA adjunct wraps in the treatment of

Page 55: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

32

osteomyelitis. Next, a critical review of gelatin matrices for growth factor delivery is

performed to elucidate potential mechanisms to sequester angiogenic growth factors for

enhanced angiogenesis. Co-electrospinning is then used to combine the candidate

antimicrobial wrap with a VEGF-loaded photo-crosslinked gelatin-methacrylate delivery

system, previously designed by our lab, to generate a substrate that controls cellular

responses and that provides bimodal release of an antimicrobial agent and VEGF. In all,

these platforms provide fundamental knowledge and tools to individually address the

complex biological processes necessary for improving the Masquelet technique in bone

loss management.

Page 56: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

33

Chapter II: Comparative Efficacy of Resorbable Fiber Wraps Loaded

with Gentamicin Sulfate or Gallium Maltolate in the Treatment of

Osteomyelitis

2.1 INTRODUCTION

Treatment of critical-sized bone defects with the Masquelet technique remains a

clinical challenge due to the high incidence of osteomyelitis associated with these type of

defects [6, 17, 163, 168, 169]. The current standard of care is superficial debridement

followed by a systemic antibiotic regimen; however, a high rate of infection recurrence

results due to inadequate debridement and limited reach of systemic antimicrobials to the

targeted tissue [106, 107, 163, 170]. Another concern with the continued use of multiple

rounds of antibiotics is the potential development of bacterial tolerance and resistance as

well as side effects [164, 171, 172]. Implant-associated infection raises additional treatment

challenges due to bacterial colonization of the biomaterial surface and formation of biofilm

that confers antibiotic resistance [173]. To increase the efficacy of the antibiotic and limit

systemic effects during the Masquelet technique, clinicians and researchers have loaded

antibiotics in the poly(methyl methacrylate) (PMMA) bone cement spacer for local

delivery [162, 174, 175]. However, there is often suboptimal release, and the well-

documented exothermic effect caused by PMMA polymerization limits which

antimicrobials can be used [36, 163, 164]. Antimicrobial coatings used as adjunct therapies

offer an alternative solution to antimicrobial-loaded PMMA spacers [173, 176]. These

coatings can be applied to the PMMA spacer by coating the surface with polymer or

Page 57: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

34

ceramic solutions containing antimicrobials [173]. Nevertheless, these coatings are often

not stable and typically require chemical modification of implants to enable stable

attachment [173]. The potential of antimicrobial coatings to migrate imposes risks that

could adversely affect treatment. Although, chemical modification of the PMMA spacer

can prevent migration, chemical modification may alter the properties of the PMMA spacer

and affect formation of the induced membrane. These limitations underscore the need for

a method to improve local antimicrobial treatment during the Masquelet technique.

Resorbable wraps are an emerging alternative that can be used broadly and provide

independently-controlled antimicrobial release kinetics [177-179]. These wraps can also

be used as stand-alone treatments to prevent infection in compound fractures that do not

require grafting. Hydrogel-based wraps, such as collagen fleece, offer promise as adjunct

therapies due to their simple fabrication and ease of application by clinicians.

Antimicrobials are loaded into the water-swollen matrix and lyophilized to encapsulate

them. Researchers have demonstrated sustained release with kinetic control provided by

modulation of the porosity or chemical crosslinking [172, 180-182]. Similarly,

polyelectrolyte multilayer films have been investigated as adjunct antimicrobial wraps for

bone defects due to their ease of fabrication, antimicrobial incorporation, and clinical

application. They are generally fabricated by layer-by-layer (LbL) assembly which enables

sequential adsorption of complementary electrostatic species to a pre-functionalized

substrate. Polyelectrolyte multilayer wraps have been reported to control release of

antimicrobials by diffusion through the various layers [123, 183]. Both hydrogel-based

wraps and polyelectrolyte multilayer wraps are able to modulate release and expand the

Page 58: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

35

collection of antimicrobials that can be incorporated, as compared to PMMA spacers.

Although the collection of antimicrobials that can be used is expanded, the aqueous

processing conditions required for these fabrication techniques limit the collection to

water-soluble antibiotics [172, 183]. Careful consideration should be given to the potential

risk of tolerance development associated with overuse of antibiotics, and the variable tissue

penetration of antibiotics in bone [134, 171, 184]. Another concern with the use of these

fabrication techniques is that the antimicrobial encapsulation efficiency is limited by

swelling-based absorption into the bulk matrix or affinity-based adsorption onto the surface

[172, 183]. Restriction of the antimicrobial load can potentially reduce the efficacy of the

treatment. These constraints warrant the investigation of alternative fabrication techniques

that enable embedment of novel antimicrobials into adjunct wraps.

Cast antimicrobial films offer an alternative solution to hydrogel and

polyelectrolyte multilayer wraps due to the broad selection of materials and ease of

fabrication. This was demonstrated by polyester-based solutions that were blended with

various concentrations of gentamicin sulfate and cast into antimicrobial films that were

designed to augment orthopedic implants. Results confirmed a high loading capacity of

gentamicin sulfate; however, processing conditions of the films resulted in high burst

release due to significant levels of surface-bound gentamicin sulfate [185, 186]. In contrast

to films, electrospinning offers notable advantages for antimicrobial wraps due to its ease

of fabrication, versatility in materials, and tunable release kinetics [179, 187].

Electrospinning generates fibrous wraps by application of an electric field to a polymer

solution loaded with antimicrobial agents. Electrospun wraps enable greater control over

Page 59: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

36

release kinetics by altering the chemical properties and fiber microarchitectures (e.g. fiber

diameter) [187]. Recent studies have established the potential of electrospun wraps to

control release of water-soluble antibiotics for treatment of osteomyelitis [188, 189]. Gao

et al. demonstrated that electrospun poly(lactic-co-glycolic) acid (PLGA) provided

controlled released of vancomycin over 4 weeks with corresponding bactericidal activity

resulting in significantly reduce bacterial density in an infected rabbit femoral defect model

after 8 weeks as compared to controls containing no vancomycin or systemic delivery of

vancomycin. However, the antimicrobial activity of the electrospun wrap was not

established with an osteomyelitis-derived strain [188]. This is important as antimicrobial

susceptibility varies among different bacterial strains [190]. This study was also limited by

the lack of evaluation of the potential cytotoxic effect of vancomycin as it relates to

establishing the safety and efficacy of the agents [188]. Similarly, Wei et al. demonstrated

sustained release of vancomycin from electrospun polycaprolactone (PCL) with released

concentrations up to 2 weeks. The wrap was confirmed to be cytocompatible with

osteoblasts and significantly decreased bacterial density in an infected rabbit femoral defect

model as compared to blank scaffolds over 12 weeks. However, the lack of relevant

osteomyelitis-derived strains used in the evaluation of treatment efficacy again limited the

potential impact of this study. The study also failed to perform a direct comparison of the

cytotoxic effects to the efficacy of the released vancomycin [169]. Furthermore, very few

studies have highlighted the capacity of electrospinning to enable loading and release of

novel antimicrobials [179].

Page 60: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

37

The purpose of this study is to perform a comparative analysis of the efficacy of

gentamicin sulfate and gallium maltolate released from resorbable electrospun wraps in the

treatment of osteomyelitis. Electrospinning parameters were identified to generate

comparable release profiles of gentamicin sulfate and gallium maltolate from electrospun

poly(lactide-co-glycolide) (PLGA) wraps. Gallium maltolate is a novel antimicrobial that

mimics the ferric ion which allows it to exploit the iron-dependence of microorganisms

and inhibit growth [191]. To date, the antimicrobial potential of gallium maltolate has been

demonstrated in chronic wound applications but not in osteomyelitis applications [141,

192]. This study provides a direct comparison of the bactericidal activity of gentamicin

sulfate and gallium maltolate by identifying the minimum inhibitory concentration and

minimum bactericidal concentration on bacterial isolates from osteomyelitis. The

antimicrobial activity was then compared to the cytotoxicity in order to establish the safety

and efficacy of each agent. Finally, in vivo bactericidal activity and bone formation was

evaluated to determine the efficacy of an antimicrobial electrospun wrap for prevention of

bone infection in critical-sized bone defects.

2.2 MATERIALS AND METHODS

Materials

All chemicals and reagents were purchased from Sigma Aldrich (Milwaukee, WI)

and used as received unless otherwise noted.

PLGA Wrap Fabrication

Page 61: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

38

50:50 PLGA (acid-terminated; inherent viscosity range 0.55–0.75 dL/g; DURECT

Corp., Cupertino, CA) was dissolved in 1,1,1,3,3,3-hexafluoro-2-propanol (HFIP)

(Halocarbon, Peachtree Corners, GA) to produce a 40% (w/v) polymer solution. Polymer

solutions were formulated with gentamicin sulfate at 1.25% of the polymer mass, gallium

maltolate (Gallixa LLC, Menlo Park, CA) at 25% of the polymer mass, or without the

incorporation of an antimicrobial agent (blank). The concentration of gentamicin sulfate

and gallium maltolate blended with the PLGA was determined by considering the release

kinetics obtained from preliminary scouting studies (data not included) and by considering

reported minimum inhibitory concentrations (MIC; lowest concentration that visibly

inhibited bacteria growth) of gentamicin sulfate and gallium maltolate [123, 141]. The

PLGA solutions were electrospun at ambient conditions (22°C, 40-50% relative humidity).

Briefly, solutions were horizontally ejected at a volumetric flow rate of 0.7 mL/h (KDS100;

KD Scientific, Holliston, MA) through an 18-gauge metal spinneret charged to 10 kV

(ES30P-5W/DDPM; Gamma High Voltage, Ormond Beach, FL). Fibers were collected on

a grounded static plate located 15 cm from the needle under ambient conditions. The

gentamicin sulfate/PLGA solution was electrospun for 4 h and the gallium maltolate/PLGA

solution was electrospun for 2 h. Meshes were then placed under vacuum for at least 24 h

prior to further characterization.

Fiber Characterization

An electrospun specimen (n= 3, 5 mm x 5 mm) was cut from the center of each

wrap (N=3/ group) to preclude edge effects. After sputter coating with 4 nm of gold

Page 62: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

39

(Sputter Coater 108, Cressington Scientific Instruments, Oxhey, Watford, UK), fiber

microarchitecture was examined with scanning electron microscopy (SEM) (Phenom Pro;

NanoScience Instruments, Phoenix, AZ) at an accelerating voltage of 10 kV. Images

captured at 500X and 2000X via raster patterning on the front and back of each specimen

were analyzed to determine representative microarchitectures. Fiber diameter was

measured from the first 10 fibers to cross the midline of each 2000X micrograph (n=900

fibers/group).

Gentamicin Sulfate Encapsulation Efficiency

The encapsulation efficiency of gentamicin sulfate in the electrospun PLGA was

determined by first dissolving PLGA mesh specimens (1 cm x 6 cm; n=3) in 2 mL of

dichloromethane (DCM). Deionized water (DI water, 2 mL) was then added to each PLGA

solution to extract the gentamicin sulfate. The aqueous extracts were frozen and lyophilized

to concentrate the gentamicin sulfate. The dose of gentamicin sulfate in each sample was

evaluated by adding 0.5 mL of 2% ninhydrin to the lyophilized product and incubating at

120C for 15 min. The ninhydrin solutions were diluted with 1 mL of DI water to enable

spectrophotometric analysis (Infinite 200 Pro, Tecan, Morrisville, NC) at 570 nm. The

encapsulation efficiency was determined based on a standard curve and the theoretical

mass of gentamicin sulfate incorporated into the samples. Blank electrospun PLGA meshes

were used as controls.

In Vitro Release of Gentamicin Sulfate

Page 63: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

40

Gentamicin sulfate release kinetics were determined by placing gentamicin sulfate-

loaded PLGA wraps (1 cm x 6 cm; n = 3) into 2 mL of DI water. DI water was selected, as

opposed to phosphate buffer, due to the low sensitivity of gentamicin sulfate in buffered

solution at the low concentrations [193]. Samples were incubated with rotation at 37C for

14 days. At daily time points, release medium was collected and replaced with fresh DI

water. The collected release medium was frozen and lyophilized to concentrate the

gentamicin sulfate. The dose released at each time point was determined using the

previously detailed ninhydrin assay. The percentage of gentamicin sulfate released daily

was calculated based on a standard curve and the initial mass of gentamicin sulfate

incorporated into each specimen. Blank electrospun PLGA meshes were used as controls.

The studies were repeated in triplicate and the results are represented in terms of the

average dose released (µg) and average cumulative release over time.

Gallium Maltolate Encapsulation Efficiency

The encapsulation efficiency of gallium maltolate in electrospun PLGA was

determined by first dissolving gallium maltolate-loaded PLGA samples (1 cm x 6 cm; n =

3) in 2 mL of DCM. Two-fold serial dilutions of the PLGA solutions were evaluated

spectrophotometrically at 322 nm to determine the dose of gallium maltolate loaded in each

sample. The encapsulation efficiency was calculated based on a standard curve and the

theoretical mass of gallium maltolate incorporated into the samples. Blank electrospun

PLGA meshes were used as controls.

Page 64: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

41

In Vitro Release of Gallium Maltolate

Gallium maltolate release kinetics were determined by placing gallium maltolate-

loaded PLGA wraps (1 cm x 6 cm; n=3) into 2 mL of DI water. Samples were incubated

with rotation at 37C for 14 days. At daily time points, release medium was collected and

replaced with fresh DI water. Gallium maltolate concentration was determined

spectrophotometrically by measuring the absorbance at 307 nm. The percentage of gallium

maltolate released daily was calculated based on a standard curve and the initial mass of

gallium maltolate incorporated into each specimen. Blank electrospun PLGA samples were

used as controls. The studies were repeated in triplicate and results are represented in terms

of dose released (µg) and cumulative release over time.

Bacteria Culture Conditions

A single methicillin-susceptible Staphylococcus aureus (MSSA) isolate harvested

from a patient with osteomyelitis (49230; ATCC®, Manassas, VA) was cultured in brain

heart infusion broth (BHIB) (Sigma-Aldrich) and incubated at 37C for 16 to 18 h. The

bacterial cells were centrifuged at 3000 x g to pellet the suspension. The inoculum was

subsequently removed and replaced with Roswell Park Memorial Institute 1640 Medium

(RPMI) (Thermo Fisher Scientific, Waltham, MA) supplemented with 1% v/v sodium

pyruvate (100 mM, Thermo Fisher Scientific) and 1% v/v GlutaMAX™ (200 mM, Thermo

Fisher Scientific). The optical density of the bacterial inoculum was analyzed

spectrophotometrically at 625 nm. Bacterial density [colony-forming-units (CFU)/mL]

was determined based on a standard growth curve. Colony counts from 10-fold serial

Page 65: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

42

dilutions of the inoculums grown on brain heart infusion agar (BHIA) (Sigma-Aldrich)

were used to confirm the bacteria density.

Antimicrobial Activity of Electrospun Wraps

The Kirby-Bauer assay was used to evaluate the antimicrobial activity of the

electrospun wraps. MSSA inoculum was suspended in supplemented RPMI at a turbidity

that matched a 0.5 MacFarland standard (1.5 x 108 CFU/mL) and then spread onto BHIA.

Electrospun PLGA wraps containing gentamicin sulfate or gallium maltolate (n = 2) were

cut into 6 mm diameter disks and placed on the inoculated BHIA. Blank electrospun PLGA

disks were used as negative controls and Whatman™ #1 paper filter disks (6 mm) loaded

with 10 µg of gentamicin sulfate or 10 mg of gallium maltolate were used as positive

controls. The positive control for gentamicin sulfate was selected in accordance with the

Clinical Laboratory Standard Institute (CLSI) standard [194]. As gallium maltolate is not

a conventional antibiotic, the positive control was selected such that the dose released

would concentrate a fixed volume of agar (0.5 mL, ~ 15 mm diameter zone) greater than a

10-fold increase of the MIC after 24 h. This control was derived from a modification of the

pharmacodynamic ratio (maximum serum concentration/ MIC ≥ 10) that is used to

determine optimal patient outcomes in clinical settings [195-197]. After incubating the

BHIA at 37 °C for 24 h, the zones of inhibition were measured for each disk. Studies were

repeated in triplicate and results are represented in terms of the average zone diameter

(mm).

Page 66: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

43

Bactericidal Activity of Gallium Maltolate

The susceptibility of MSSA to gallium maltolate was determined using a

microdilution assay according the guidelines of the CLSI [198]. Briefly, gallium maltolate

was dissolved in supplemented RPMI at a concentration of 16,000 M. Subsequent

dilutions were prepared from the sterile filtered gallium maltolate stock in supplemented

RPMI. In a 96-well plate, a 2-fold dilution of each concentration (n=3) was obtained using

the bacteria inoculum to yield final concentrations of gallium maltolate ranging from 2000

M to 8,000 M, each with an initial bacteria density of 5 x 105 CFU/mL. Positive and

negative controls consisted of sterile supplemented RPMI and untreated bacteria,

respectively. The initial bacteria density was measured spectrophotometrically at 625 nm

and was confirmed by colony counting using 10-fold serial dilutions of the negative control

bacterial inoculum. The well plate was incubated at 37 °C for 16-18 h. A quantitative

method to determine the MIC was developed for this study, as visual determination can

vary across observers. This method defines the MIC as the lowest concentration that

inhibits bacterial growth such that the treated bacteria density (CFU/mL) is not statistically

different from the initial bacteria density. Colony counting using 10-fold serial dilutions of

the treated bacterial inoculums confirmed the MIC. The minimum bactericidal

concentration (MBC) of gallium maltolate was also evaluated quantitatively and was

determined to be the concentration at which the treated bacteria density was decreased by

99.9% as compared to the initial bacteria density [199]. The MIC and MBC for gentamicin

sulfate were validated using the same methods detailed above. Briefly, a stock solution of

gentamicin sulfate was dissolved at a concentration of 6 M in supplemented RPMI. Final

Page 67: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

44

concentrations of gentamicin sulfate tested ranged from 0.5 M to 2.5 M. Studies were

repeated in triplicate and results are represented in terms of the average CFU/mL for each

concentration.

Differentiation of MC3T3-E1 Cells

The pre-osteoblastic cell line MC3T3-E1 (CRL-2593™; ATCC®, Manassas, VA)

was selected to assess cytotoxicity of gentamicin sulfate and gallium maltolate due to its

capacity to differentiate into osteoblasts and osteocytes. Briefly, MC3T3-E1 cells were first

expanded in alpha minimum essential media (α-MEM) (Caisson Lab, Smithfield, UT)

growth media, supplemented with 10% fetal bovine serum (Atlanta Biologicals, Flowery

Branch, GA) and 1% of penicillin-streptomycin (10,000 U/mL, Thermo Fisher Scientific)

at 37C. Media was replaced every 2-3 days until cells were near 80-90 % confluence. The

growth media was replaced with osteogenic media [growth media, supplemented with 10

mM β-glycerolphosphate (Sigma Aldrich), 50 μg/mL L-Ascorbic Acid (Sigma Aldrich),

and 10 nM dexamethasone (Sigma Aldrich)] to allow for differentiation. Osteogenic media

was replaced with fresh media every 2-3 days over 14 days.

IC50 and Selectivity Index for MC3T3-E1 Cells

Differentiated MC3T3-E1 cells (passage 2-6) were cultured in a 48-well plate at a

density of 25,000 cells/cm2 and incubated at 37C for 24 h. A cell viability assay was

performed on the cells to determine the inhibitory effect (IC50) and calculate the selectivity

index (IC50/MIC) of gentamicin sulfate and gallium maltolate. Briefly, gentamicin sulfate

Page 68: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

45

and gallium maltolate were dissolved in osteogenic media to produce stock concentrations

at 50,000 M and 15,000 µM, respectively. A more concentrated gallium maltolate stock

solution was not used due to solubility in the media. Sterile-filtered gentamicin sulfate and

gallium maltolate stocks were further diluted in osteogenic media to yield final

concentrations ranging from 0.5 M to 50,000 M and 25 M to 15,000 M, respectively.

Osteogenic media on differentiated MC3T3-E1 cells was replaced with the gentamicin

sulfate or gallium maltolate treatments (n =3/concentration) and cultured for 24 h. Positive

and negative controls consisted of cells grown on tissue culture polystyrene (TCPS) with

osteogenic media replaced with fresh media or 70% ethanol, respectively. To determine

cell viability, resazurin (Sigma-Aldrich) was diluted in osteogenic media according to the

manufacturer’s instructions and incubated with the cells at 37C for 4 h. Fluorescence was

measured spectrophotometrically with excitation set to 544 nm and emission set to 590 nm.

Cell viability was then calculated by normalizing the antimicrobial treatments to the

positive control. Using an embedded algorithm for “dose-response inhibition” in GraphPad

Prism 8, the relative IC50 value was generated. Studies were repeated in triplicate with data

presented in terms of the average viability for each concentration. The IC50 was then used

to calculate the selectivity index (IC50/MIC).

Statistical Analysis

Data averages are accompanied with ± standard deviation. Statistical analysis was

performed utilizing a standard one-way ANOVA with Tukey’s post-hoc analysis unless

otherwise indicated in figure captions. Statistical significance was accepted at P<0.05.

Page 69: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

46

2.3 RESULTS

Antimicrobial Loading and Release

Previous studies have demonstrated the ability of electrospinning to incorporate and

tune release of a variety of drugs. In this current study, we utilized electrospinning to

fabricate antimicrobial wraps loaded with gentamicin sulfate or gallium maltolate (Figure

2.1A). Electrospun PLGA was loaded with gentamicin sulfate or gallium maltolate, with a

blank wrap used as the control. The extent of antimicrobial encapsulation in each PLGA

wrap was determined by evaluating the loading efficiency. A loading efficiency of 96.9 ±

2.8% was calculated for gentamicin sulfate and 99.4 ± 12.5% for gallium maltolate. These

high efficiencies indicated that electrospinning enabled significant incorporation of two

distinct antimicrobials. The average fiber diameter of the gentamicin sulfate-loaded wrap

was 2.02 ± 0.44 µm and was 1.89 ± 0.34 µm for the gallium maltolate-loaded wrap (Figure

2.1B & C). The wraps were fabricated with similar fiber diameters in order to isolate the

effect of release kinetics for each antimicrobial, as differences in fiber diameter can

influence the diffusion path length and resorption period.

Page 70: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

47

Figure 2.1: Fabrication of antimicrobial wraps. A) Schematic of electrospinning

apparatus and scanning electron micrographs of electrospun PLGA fibers loaded with B)

gentamicin sulfate and C) gallium maltolate. Main micrograph scare bar = 100 µm. Inset

micrograph scare bar= 30 µm.

Page 71: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

48

Release kinetics were subsequently investigated after confirming high loading

efficiencies. Studies conducted in DI water demonstrated release profiles with similar burst

releases of 33.2 ± 2.6% from gentamicin sulfate-loaded wraps and 27.3 ± 1.7% from

gallium maltolate-loaded wraps. However, cumulative release after 14 days was lower for

gentamicin sulfate-loaded wraps than for gallium maltolate-loaded wraps (Figure 2.2).

This was possibly due to the higher loading fraction of gallium maltolate resulting in faster

release kinetics as compared to the gentamicin sulfate. Despite these differences, the daily

dose of gentamicin sulfate and gallium maltolate released were greater than the respective

MICs reported for S. aureus over 72 h (Figure 2.2) [123, 141]. This data demonstrated that

electrospun PLGA wraps enable the incorporation and controlled release of a diverse

portfolio of antimicrobials at therapeutic concentrations.

Page 72: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

49

Figure 2.2: Evaluation of in vitro release kinetics from electrospun antimicrobial PLGA

wraps in DI water. Cumulative release and daily release of A) gentamicin sulfate and B)

gallium maltolate was evaluated over 2 weeks. Red-dashed line indicates hypothesized

MIC for each antimicrobial.

In Vitro Bacterial Inhibition and Cellular Response

To confirm the antimicrobial activity of the electrospun PLGA wraps, the Kirby

Bauer diffusion disk assay was used. The positive gentamicin sulfate control resulted in a

zone of inhibition of 17.9 ± 0.6 mm while the negative control did not inhibit growth, as

expected. Gentamicin sulfate released from PLGA wraps formed a zone of inhibition

Page 73: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

50

comparable (17.8 ± 0.3 mm) to that of the positive control (Figure 2.3A). These results

indicated that the MSSA was susceptible to gentamicin sulfate and that electrospinning did

not impede gentamicin sulfate antimicrobial activity. Furthermore, the positive gallium

maltolate control did not form a zone of inhibition. This was unexpected as the control was

loaded based on the pharmacodynamic ratio (maximum serum concentration/MIC ≥10)

commonly used to predict clinical efficacy. Accordingly, released gallium maltolate did

not form a zone of inhibition (Figure 2.3B). The lack of zone formation suggested that

MSSA was not susceptible to gallium maltolate at the hypothesized MIC concentration.

Page 74: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

51

Figure 2.3: Kirby Bauer assay was used to evaluate bioactivity of the antimicrobial

wraps. Representative images of the inhibition zones in response to A) gentamicin sulfate

and B) gallium maltolate released from the PLGA wrap (bottom half images), as

compared to negative control (blank PLGA wrap, top right image) and the positive

control (solubilized gentamicin sulfate or gallium maltolate, top left image) after 24 h.

Graphs display the corresponding measurements of zone diameters. * indicates statistical

differences with respect to the gentamicin sulfate positive control (P<0.05).

Page 75: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

52

To determine the antimicrobial susceptibility of MSSA to gentamicin sulfate and

gallium maltolate, a microdilution study was performed. The number of CFUs resulting

from the treatments after 24 h was used to determine the MIC and MBC for each

antimicrobial. The actual MIC of gentamicin sulfate was 1.0 µM and was found to be

higher than the hypothesized MIC reported by Moskowitz et al. [123]. Despite the MIC

being higher than expected, release kinetics provided daily release greater than this

concentration. The MBC was found to be 1.5 µM and was also within the range of

concentrations released daily, confirming that gentamicin sulfate is able to both inhibit

growth and kill MSSA at concentrations released from the PLGA wrap (Figure 2.4A). The

actual MIC of gallium maltolate was 6000 µM and was also discovered to be higher than

the MIC reported by Cereceres et al. (Figure 2.4B) [141]. The MBC was not assessed as

it exceeded the solubility limit in RPMI. Although these results confirm that gallium

maltolate is able to inhibit growth of MSSA, the PLGA wrap does not release

concentrations high enough to effectively eradicate MSSA infection.

Page 76: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

53

Figure 2.4: The evaluation of the MIC and MBC of A) solubilized gentamicin sulfate

and B) solubilized gallium maltolate on MSSA bacterial colony growth after 24 h. The

mean initial bacteria density is denoted by the red dashed-line. The MIC was deemed the

lowest concentration that inhibited bacterial growth such that the treated bacteria density

(CFU/mL) is not statistically different than the initial bacteria density. * indicates

statistical differences with respect to the initial bacteria density, (P<0.05). The blue

dashed-line represents a 99.9% reduction in the initial bacteria density. The MBC was

deemed the lowest concentration that reduced bacterial growth ≥99.9% of the initial

bacteria density.

The effect of gentamicin sulfate and gallium maltolate on differentiated MC3T3-

E1 cell viability was also investigated to determine the selectivity indices. The selectivity

index is a ratio of the MIC to IC50 that used to establish the efficacy and safety of an

antimicrobial agent. The relative IC50 of gentamicin sulfate was 566.5 ± 142.4 µM and

778.6 ± 326.1 µM for gallium maltolate (Figure 2.5A & B). The corresponding selectivity

indices (IC50/MIC) were 566.5 ± 142.4 and 0.1± 0.1, indicating that gentamicin sulfate is

a safer and more effective antimicrobial than gallium maltolate for treating MSSA

infection. As such, the gentamicin sulfate-loaded PLGA wrap was selected for further in

vivo evaluation.

Page 77: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

54

Figure 2.5: Viability of differentiated MC3T3-E1 cells relative to TCPS control after 24

h exposure to various concentrations of A) solubilized gentamicin sulfate and B)

solubilized gallium maltolate. Red-dashed line indicates relative IC50 calculated using

GraphPad Prism 8. A relative IC50 was identified as 566.5 ± 142.4 µM for gentamicin

sulfate and 778.6 ± 326.1 µM for gallium maltolate.

2.4 DISCUSSION

The observed recurrence of osteomyelitis during treatment of the Masquelet

technique and the risk of implant-associated infection in critical-sized bone defects created

a clear need for a resorbable device that offered local antimicrobial delivery. We sought to

develop an electrospun antimicrobial wrap that could be used broadly as a stand-alone

treatment or as an adjunct treatment to prevent infection. Electrospinning was selected as

it allows higher antimicrobial encapsulation by blending antimicrobials with the polymer,

as compared to other wrap fabrication techniques that rely on swelling-based absorption or

surface adsorption. Furthermore, electrospinning enables tunable release of antimicrobials

by modulating chemical properties and fiber microarchitectures [200, 201]. Antibiotics are

traditionally selected as the antimicrobial agent incorporated into electrospun wraps as they

are effective against a wide spectrum of bacterial species making them useful for various

applications. They attack bacteria by disrupting a specific biological process such as cell

wall synthesis, nucleic acid synthesis, or protein synthesis [134, 202]. Although antibiotics

are effective at preventing infection, overuse of them can lead to bacterial modifications

that increase efflux, decrease binding, or cause inactivation of the antibiotics [202]. This

risk has prompted the investigation of novel antimicrobials like metals. Metal compounds

are of interest due to the ability to target multiple biological processes as opposed to the

Page 78: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

55

single-target approach of antibiotics [134]. There are few studies that have investigated the

efficacy of metal compounds as an alternative solution to antibiotics in the treatment of

osteomyelitis.

To this end, we compared the efficacy of gentamicin sulfate and gallium maltolate

released from electrospun PLGA wraps in the treatment of osteomyelitis. PLGA was

selected as the polymer due to its established use as a controlled delivery system for low

molecular weight antimicrobials [185, 203, 204]. Modification of the co-polymer ratio,

molecular weight, or chemical end-groups can be used to tune release kinetics [205].

Gentamicin sulfate was chosen as the antibiotic owing to its broad antimicrobial spectrum

and frequent clinical use in bone infection [123]. The mode of action of gentamicin sulfate

involves inhibition of protein synthesis by targeting the 30S or 50S subunit of bacterial

ribosomes [202]. Furthermore, the potential to overcome antibiotic resistance reinforced

the selection of gallium maltolate as a metal-based alternative to gentamicin sulfate. The

atomic similarity to ferric iron improves bacterial uptake of gallium maltolate. Once taken

up by bacteria, the inability to reduce gallium ions disrupts the iron-redox process that is

responsible for iron transport, respiration, nucleic acid synthesis, and reactive oxygen

species protection leading to death [141, 206]. The concentration of gentamicin sulfate and

gallium maltolate blended with the PLGA was selected based on predicted release kinetics

of the PLGA wrap. It was postulated that the release kinetics would result in daily release

greater than the hypothesized MIC concentration for each antimicrobial over at least 72 h,

which is the critical period when vulnerable planktonic bacteria can progress into a resistant

biofilm [123, 141, 207, 208]. HFIP was selected as the electrospinning solvent, as organic

Page 79: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

56

solvents enable solubilization of gallium maltolate at high concentrations [141]. Although

gentamicin sulfate is moderately soluble in HFIP, achieving release of the hypothesized

MIC does not require a high loading concentration as compared to gallium maltolate. It

was consequently determined that utilizing HFIP to solubilize both antimicrobials was

acceptable. HFIP evaporation during electrospinning ensured direct encapsulation of each

antimicrobial resulting in high encapsulation efficiencies. A high burst release was

expected to occur in response to the incompatible interactions between the hydrophilic

drugs and hydrophobic PLGA [203, 204]. This is of particular concern for analysis of

gentamicin sulfate release kinetics. Given the low loading concentration, a high burst

release reduces the amount of drug available at later time points during the sustained release

phase. The sustained release doses may fall below the detection limit of the ninhydrin assay

and prevent analysis. Therefore, the gentamicin sulfate-loaded wrap was electrospun for a

longer period to increase the thickness of the wrap and the corollary amount of drug

available during the sustained release phase. Although the wraps were fabricated with

different thicknesses, the fiber diameters were comparable and enabled normalization of

the release kinetics. The release profiles were expected to be comparable due to similar

drug-polymer interactions. However, the gallium maltolate-loaded wrap exhibited higher

cumulative release beyond the burst phase. It was hypothesized that the differences in

release kinetics were due to the loading concentration of each drug. Drugs loaded above a

polymer solubility threshold have the potential to aggregate into clusters in the amorphous

region of semi-crystalline polymers. As the aqueous medium penetrates the matrix, these

clusters are released via dissolution resulting in faster cumulative release than an evenly

Page 80: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

57

dispersed drug load. This phenomenon has been demonstrated with other electrospun

polyester (i.e. polycaprolactone) meshes loaded with various concentrations of drugs [209,

210]. It is possible that the high loading fraction of gallium maltolate resulted in cluster

formation in the amorphous region of PLGA leading to faster release kinetics as compared

to the gentamicin sulfate-loaded wrap. Despite these differences, both wraps were able to

provide release of the respective antimicrobial above its hypothesized MIC in the first 72

h [207, 208]. These findings confirmed that electrospinning enables encapsulation and

release of different types of antimicrobials over a clinically relevant duration.

A potential concern with direct encapsulation of bioactive agents during

electrospinning is a loss of biological activity. The antimicrobial activity of each wrap was

evaluated using the Kirby Bauer assay which is a standardized assay for evaluation of

antimicrobial susceptibility [194]. MSSA was designated as the testing strain for its

prevalence in osteomyelitis [211]. The gentamicin sulfate-loaded wrap demonstrated

bioactivity comparable to the positive control indicating that the MSSA is susceptible to

gentamicin sulfate and that electrospinning does not impede the antimicrobial activity. As

gallium maltolate is not a conventional antibiotic, a standard has not been determined for

evaluating susceptibility. We selected the gallium maltolate standard based on a

modification of a pharmacodynamic ratio (maximum serum concentration/MIC ≥10) that

is commonly used to predict optimal clinical outcomes and suppression of resistance [195,

196]. Rather than considering a maximum serum concentration, a maximum agar

concentration in a constant volume of agar was considered. It was assumed that complete

release of the gallium maltolate from the paper filter disk would occur over 24 h and

Page 81: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

58

concentrate the agar above the hypothesized dose based on uncontrolled diffusion from the

paper-filter disk [194]. However, the gallium maltolate control did not form a zone of

inhibition suggesting that MSSA was not susceptible to gallium maltolate at the

hypothesized MIC. This MIC was targeted based on the reported activity of gallium

maltolate against MRSA [141]. It is well known that antimicrobial activity is strain-

dependent so this data was not unforeseen [190]. We evaluated the susceptibility of MSSA

to soluble gallium maltolate to identify the MIC using a microdilution assay. Results

indicated that MSSA was susceptible to gallium maltolate at higher concentrations than

those reported for MRSA and that the current iteration of the PLGA wrap does not release

efficacious concentrations of gallium maltolate. Similarly, the MIC of gentamicin sulfate

was found to be higher than what was reported for MSSA. This was expected as Moskowitz

et al. evaluated a bacteria density below (5-fold) the CLSI standard density that was used

in present study [123]. It is probable that a low bacteria density will be susceptible to an

antimicrobial at a relatively low concentration as compared to a high bacteria density.

Despite this finding, the PLGA wrap was able to release daily concentrations above the

actual MIC of gentamicin sulfate.

To determine the ability of gallium maltolate to selectively target MSSA while

retaining cell viability of MC3T3-E1 cells at this relatively high MIC, we evaluated the

selective bioactivity as compared to gentamicin sulfate. A comparative analysis of the two

agents revealed a significantly lower selectivity index for gallium maltolate compared to

gentamicin sulfate. This indicated that the efficacy of gallium maltolate is undermined by

its cytotoxic effect on osteogenic cells and therefore should not be used to treat

Page 82: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

59

osteomyelitis. Despite these findings, gallium maltolate-loaded PLGA wraps should be

further explored for controlled delivery in other biomedical applications such as chronic

wounds [141, 212]. Further evaluation of the efficacy of the gentamicin sulfate-loaded

PLGA wrap in the treatment of osteomyelitis is currently in progress in an infected rat

segmental defect model.

Overall, the gentamicin sulfate-loaded PLGA wrap utilizes material properties and

fibrous microarchitecture to maintain release in a controlled manner. Coupled with the

superior in vitro and in vivo antimicrobial performance, this wrap demonstrates a strong

potential as an adjunct therapy in the treatment of osteomyelitis associated with open

fractures and implant-mediated colonization. There were limitations in the present study

that necessitate future research. Current studies are in progress to evaluate the in vivo

antimicrobial activity of the gentamicin sulfate-loaded PLGA wrap. However, further

analysis is needed that provides a rigorous in vivo assessment of efficacy in bone

regeneration of infected defects. These studies will need to investigate bone volume

fraction, defect bridging, and total bone growth within the defect following treatment.

Results would be compared to antibiotic-loaded cement in order to assess improvement

upon clinical standards. In all, the gentamicin sulfate-loaded PLGA wrap has the potential

to have a direct impact on orthopedic applications through rational design of adjunct

infection control combined with the capacity to enhance fracture healing.

Page 83: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

60

2.5 CONCLUSIONS

In the present study, we successfully performed a comparative analysis of efficacy

of two antimicrobial-loaded, resorbable meshes for the treatment of osteomyelitis during

the Masquelet technique. Our analysis resulted in the selection of an antimicrobial wrap

that sustains release of an antibiotic with established bactericidal activity for the

osteomyelitis bacterial strain. Specifically, the completed studies establish an initial proof-

of-concept wrap with the ability to reduce infection for potential use as an adjunct therapy

for contaminated fractures and implants. This wrap has been optimized for sustained

release to ensure bactericidal concentrations are retained throughout the treatment. The

ability of this treatment to selectively target bacteria associated with osteomyelitis while

avoiding cytotoxicity of osteoblasts was also evaluated to establish the potential safety and

efficacy of this treatment. Future work will build upon the present research by evaluating

fracture healing with the electrospun gentamicin sulfate-loaded PLGA wrap in a

contaminated femoral defect model.

Page 84: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

61

Chapter III: Gelatin Matrices for Growth Factor Sequestration 1

3.1 POLYMERIC MATRICES FOR GROWTH FACTOR DELIVERY

The bioactive potential of the induced membrane is heavily regulated by the

vasculature; therefore, the temporal decrease in the vasculature observed during treatment

imposes significant limitations on the success of the Masquelet technique [15, 16, 21, 22].

Overcoming these limitations will require an increase the vasculature at later stages. This

can be achieved by implementing a strategy to enhance angiogenesis in the induced

membrane. One of the most common strategies investigated for enhanced angiogenesis in

tissue engineering applications is growth factor delivery. Angiogenic growth factors

instruct cellular responses such as cell migration, proliferation, and differentiation during

tissue repair and remodeling [72, 74, 213]. The ability of growth factors to direct cellular

behavior is dependent on the concentration as well as the spatial dispersion. Bolus delivery

of growth factors display limited efficacy and adverse side effects such as ectopic growth

and carcinogenic effects. Researchers attempt to address these limitations by developing

materials to provide localized delivery and controlled release [60].

Advances in polymeric material design over the last 25 years have enabled the

development of tunable platforms for growth factor delivery [214-216]. Synthetic

polymers (e.g. poly(lactide-co-glycolide) (PLGA), polylactide (PLA), polyglycolide

1 This work was reprinted with permission from “Gelatin matrices for growth factor sequestration”, by

Taneidra Walker Buie, Joshua McCune, and Elizabeth Cosgriff-Hernandez. Trends in Biotechnology,

2020, 38(5), 546-557. © 2019 Elsevier Ltd. All rights reserved.

Page 85: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

62

(PGA), polycaprolactone (PCL)) offer several advantages including ease of manufacture,

tunable degradation, and established use in small molecule delivery [58]. However, the

harsh processing conditions required for fabrication of synthetic polymers, such as high

temperatures or organic solvents, can denature growth factors leading to a loss in

bioactivity [59]. To circumvent this loss of bioactivity due to processing, growth factors

can be loaded into the matrix after fabrication. Post-fabrication loading can restrict the

loading capacity to adsorption to the surface or absorption in the water-swollen polymer

matrix [60]. In addition, degradation of synthetic polymers can result in an inflammatory

response due to toxic by-products or changes to the local pH [58]. As an alternative, natural

polymers and their derivatives, such as collagen, gelatin, chitosan, and alginate, are often

processed in aqueous solvents. These mild processing conditions allow for in-line loading

of the growth factors with a corollary increase in loading capacity over synthetic matrices.

As biological materials, degradation byproducts are cytocompatible and readily cleared

from the body [61, 217]. One of the more common natural polymers used for growth factor

delivery is gelatin due to its versatile fabrication processing, ease of modification, and its

electrostatic properties that confer growth factor affinity [218, 219]. There has been an

increase in the development of gelatin delivery systems that provide tunable delivery of

growth factors to support bioactivity retention [220-222]. However, sequestration and

release is primarily governed by an increase in the crosslink density resulting in structural

changes to the gelatin matrix [63]. The focus of this review is to provide a summary of

alternative mechanisms to enhance growth factor sequestration in gelatin matrices, current

Page 86: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

63

gelatin growth factor matrices in practice, and future perspectives of gelatin matrices in

tissue engineering.

3.2 AFFINITY SEQUESTRATION TO CONTROL GROWTH FACTOR RELEASE

The efficacy of growth factor therapy for tissue engineering applications is highly

dependent on retaining the bioactivity during fabrication and application. Gelatin matrices

offer advantages over synthetic polymeric carriers due to its mild fabrication conditions

(e.g. aqueous solution processing) and high growth factor loading during fabrication [223,

224]. Standard processing of collagen to generate gelatin also increases its solubility and

provides ease of fabrication as compared to collagen delivery vehicles. The selected

hydrolytic treatment (acidic or basic) determines the isoelectric point (IEP) of gelatin

matrices, the pH at which the charge on the gelatin is zero [225-227]. Acidic pre-treatment

results in positively-charged gelatin (type-A gelatin) with an IEP between pH 8-9. Alkaline

pre-treatment hydrolyzes amide residues to carboxyl residues leading to negatively-

charged gelatin (type-B gelatin) with an IEP between pH 4.8-5.4 [228]. The net charge of

gelatin enables electrostatic interactions with oppositely charged growth factors which

inherently sequesters growth factors. However, rapid dissolution of gelatin during

implantation requires gelatin matrices to be crosslinked into hydrogels [229]. The

crosslinking modality and degree of crosslinking can strongly impact the resulting physical

properties of the gelatin matrix [230, 231]. Reagent-based crosslinking modalities enable

homogenous crosslinking and are categorized as either non-zero length or zero-length

Page 87: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

64

based on assisted bonding or direct bonding, respectively [232, 233]. Non-zero length

crosslinking reagents (e.g. aldehydes, isocyanates, and polyepoxides) react with free amine

residues and/or carboxylic acid residues to form intramolecular and intermolecular

crosslinks within a gelatin solution. Zero-length crosslinking agents (e.g. acyl azides and

carbodiimides) facilitate the direct reactions between carboxylic acid residues and amine

residues on the same gelatin molecule or adjacent gelatin molecules without intermediate

molecules in the network [234]. However, these chemical crosslinking modalities can have

residual unreacted reagents that could compromise the biocompatibility of the gelatin

matrices. Less toxic, covalent crosslinking modalities include natural enzymes such as

genipin, which is a natural reagent derived from the gardenia fruit. It facilitates crosslinking

in a two-step process that first reacts with amine residues of gelatin followed by reaction

with esters of genipin with amine residues of gelatin [229]. Photo-polymerization is another

common method of covalently crosslinking gelatin. This method requires functionalization

of gelatin with a primer (e.g. acrylamide, methacrylamide) in order to undergo photo-

polymerization in the presence of free radicals [235]. Use of these modalities offer versatile

methods for controlling the mechanical and physical properties of gelatin matrices.

Page 88: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

65

Figure 3.1: The degree of crosslinking affects the hydrogel mesh size that governs

growth factor release from gelatin matrices. A) Low crosslinking results in rapid swelling

and diffusion. B) High crosslinking results in reduced swelling and sustained diffusion.

The crosslink density determines the mesh size of the hydrogel, which is a primary

consideration in the sequestration and release of growth factors in gelatin matrices (Figure

3.1). Growth factors that are smaller than the effective mesh size diffuse out rapidly and

are at risk for proteolytic degradation; whereas, growth factors that are larger than the

effective mesh size are sequestered and protected. As such, modulation of the mesh size by

changing the gel crosslink density provides a mechanism to tune the release profile.

Crosslink density also affects a number of gel physical properties including swelling,

mechanical properties, and degradation rate [63]. Growth factor conjugation has been

investigated as an alternative to sequester growth factors irrespective of the hydrogel mesh

Page 89: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

66

size. As mentioned previously, gelatin contains several chemical groups that enable

covalent crosslinking within gelatin or to adjacent gelatin molecules. These chemical

reactions can covalently bind growth factors to gelatin to enhance sequestration [236-238].

Release of conjugated growth factors will be delayed until cleavage of gelatin matrices

and/or linkers permit diffusion (Figure 3.2) [239].

Figure 3.2: Effect of conjugation on growth factor sequestration in gelatin matrices.

Growth factor-conjugated gelatin matrix displays burst release due to initial swelling that

releases non-conjugated growth factors followed by sustained growth factor release after

proteolytic chain scission.

Among these conjugation modalities are bi-functional crosslinkers such as

diisocyanates or susuccinimidyl valerate which facilitate covalent bonding with the

available free amines on gelatin and growth factors [239, 240]. However, it is possible for

side reactions to occur such as a single bi-functional crosslinker binding two growth factors

or multiple bi-functional crosslinkers binding a single growth factor due to the ratio of

amines present of growth factors [240]. The latter could affect the hydrogel mesh size by

behaving as an additional crosslink point with the gelatin. Alternatively, a two-step process

Page 90: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

67

of functionalization of the gelatin and growth factor independently following by a

conjugation step provides additional control over the reaction. One of the most common

examples of this process is the use of methacrylated gelatin and acrylated or methacrylated

growth factors that undergo free radical polymerization in the presence of a photo-initiator

and UV irradiation [241]. Although conjugation modalities have been successful at

immbolizing growth factor for sustained sequestration, the poor control of conjugation sites

on growth factors, typically non-specific amino groups, puts these techniques at a high

potential for bioactivity loss [242]. To address this limitation, affinity sequestration has

been explored as a means to sequester the growth factor for sustained release without loss

of bioactivity and minimal effect of the gelatin physical properties. Common affinity

sequestration approaches will be described in detail with a focus on the relationships with

physiochemical properties and gelatin matrix design, Figure 3.3.

Figure 3.3: Overview of the physiochemical properties governing growth factor diffusion

from gelatin matrices. The properties included growth factor affinity to A) ligands, B)

adaptor proteins, and C) nanomaterial additives incorporated into gelatin matrices.

Page 91: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

68

The established interactions of growth factors and the extracellular matrix (ECM)

is a rich field to draw design inspiration for sequestering growth factors. As such, ECM-

derived ligands are one of the most common moieties used to sequester growth factors in

gelatin matrices. These non-covalent bonds do not impair the stability or bioactivity of

growth factors [64, 65, 242]. Among these target ECM ligands is heparin, a negatively-

charged glycosaminoglycan that has binding domains for several growth factors. Heparin

binds several growth factors via electrostatic interactions between amino acid residues and

carboxyl groups. It has commonly been incorporated into gelatin through functionalization

by 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride/N-

hydroxysulfosuccinimide (EDC/NHS). EDC/NHS reactions facilitate crosslinking

between carboxyl groups on heparin and amino acid residues on gelatin, but intramolecular

or intermolecular crosslinking of gelatin is also possible with this technique [65, 243]. This

can be avoided by first activating the carboxyl groups on heparin with EDC prior to its

incorporation into gelatin [244]. The heparin-modified gelatin matrix can then be used to

sequester growth factor with high affinity and without chemically modifying the growth

factor. An adapter protein is another type of ligand-based moiety that is composed of a

coiled peptide and a collagen-binding domain (CBD) derived from fibronectin. CBDs

derived from fibronectin have high affinity towards collagen and gelatin and facilitates

ready modification of gelatin matrices [64]. The coiled peptide tethered to the CBD enables

electrostatic binding with a complementary coiled peptide tethered to a growth factor of

interest. This technology has proven to be highly adaptable and can be modified to increase

the binding strength to gelatin by altering the source of CBD [64]. Furthermore, high-

Page 92: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

69

throughput screening of DNA/RNA libraries using systematic evolution of ligands by

exponential enrichment (SELEX) technique enables selection of peptide and

oligonucleotide aptamers with high binding affinity by electrostatic interactions [66, 245].

Aptamers can be incorporated into gelatin matrices by standard bioconjugation methods

[66]. As an alternative to chemical modification of the gelatin matrix, nanomaterial

additives with growth factor affinity have been explored to generate gelatin nanocomposite

delivery systems. These additives can be readily mixed into gelatin precursor solutions and

provide a high surface area to facilitate growth factor adsorption. Common nanomaterials

used to sequester growth factors in gelatin matrices are nanodiamonds, carbon-based

nanoparticles with truncated octahedral structures, and nanoclays. Functional groups on

the surface of nanodiamonds determine interfacial interactions with growth factors. These

interaction include electrostatic, hydrogen bonds, dipole-dipole, and hydrophobic

adsorption and vary depending on the processing technique used during the synthesis of

the nanodiamonds [246]. Surface modification of the nanodiamonds through carboxylation

or hydroxylation can also be used to provide covalent conjugation to gelatin prior to

crosslinking of gelatin for greater stability [67, 246]. As compared to other widely used

carbon-based nanomaterials such as graphene oxide and carbon nanotubes, nanodiamonds

have greater biocompatibility [246]. Two-dimensional nanoclays are another type of

nanomaterial with superior biocompatibility. Nanoclays are discs composed of an

octahedral sheet of magnesium oxide inserted between two parallel tetrahedral sheets of

silica which results in negatively-charged surfaces and a positively-charged edge. Sodium

ions adsorbed to the surface of nanoclays during manufacturing foster ionic interactions

Page 93: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

70

with neighboring nanoclays in dry environments. However, nanoclays dissociate in ionic

aqueous solutions due to favorable interactions between the sodium ions and hydroxide

molecules or other ions. Dissociation allows for rearrangement and greater access to the

charged surfaces by proteins [68]. Similar to nanodiamonds, nanoclays are generally

incorporated into gelatin solutions prior to crosslinking [68, 247].

There are several applications in tissue engineering that have used these affinity

sequestration approaches to achieve growth factor delivery ranging from 5 days to 25 days

including cardiovascular repair [247], angiogenesis [48, 66, 69], bone healing [65], and

wound healing [64, 248]. For example, adaptor proteins with coiled-CBDs specific for

gelatin were incorporated into EDC/NHS-crosslinked gelatin. Epidermal growth factor

(EGF) tethered with a complementary coil was added to the gelatin to allow for non-

covalent binding. This non-covalent binding resulted in sequestration of EGF for over four

days [64]. Given this relatively moderate time frame, this matrix could be employed in

wound healing to initiate cell proliferation for tissue regeneration. Alternatively, the strong

binding affinity of heparin to vascular endothelial growth factor (VEGF) has been used in

a gelatin composite wrap that was crosslinked with EDC/NHS. This matrix was able to

sustained release of VEGF over three weeks rendering it useful to direct capillary

formation, homogenization, and maturation of blood vessels that typically occurs during

the first three weeks of angiogenesis [48]. In addition to using these modalities for

moderate sequestration, they can be selected for high specificity to increase the time that

growth factors are preserved in gelatin matrices. This is especially true for aptamers as

demonstrated in a study that selected the acrydite oligonucleotide using SELEX due to its

Page 94: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

71

high bind specificity for VEGF [66]. Another advantage of these affinity sequestration is

that they can be used for concentrated localization of growth factors to a particular area in

the matrix. For example, a single nanodiamond can bind multiple growth factors based on

its high surface area that allows for increased adsorption [246]. Furthermore, spatial

concentration of nanodiamonds in gelatin matrices provides another mean to sequester

growth factors by regulating the diffusion path length as later discussed. Despite the

potential to sequester growth factors with minimal impact on the bioactivity, careful

consideration must be given to the transient and reversible interactions that govern

sequestration when sustained preservation is desired. Another consideration is that freely

encapsulated nanomaterials can potentially bind the gelatin matrix due to their high surface

area resulting in changes to the physical properties [69].

3.3 GELATIN MATRICES IN TISSUE ENGINEERING

An accompanying aspect that affects growth factor release kinetics is the

fabrication technique, which determines the diffusion path length through the gelatin

matrix [249]. Advancements in fabrication of gelatin-based matrices have provided several

opportunities to create systems for the controlled delivery of growth factors. The geometry

and size of gelatin matrices can be altered through various fabrication techniques to control

growth factor sequestration. These strategies are primarily focused on changing the

surface-area-to-volume ratio and the diffusion path length of the embedded growth factors

[250, 251]. As a general consideration, an increase in surface area will decrease the

diffusion path length with a corollary increase in the release kinetics. These considerations

Page 95: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

72

can be applied to the design of the gelatin delivery system regardless of the resulting

geometry (e.g. microparticles, gels, fibrous meshes). The following section will describe

fabrication techniques that control growth factor sequestration for growth factor delivery

in a variety of tissue engineering applications.

3.3.1 Gelatin Microparticles

Gelatin microparticles offer several advantages as a growth factor delivery vehicle

such as a high surface area-to-volume ratio. Typically, smaller microparticles display faster

growth factor release rates due to an increase in surface area and a shorter diffusional path

length of embedded growth factors (Figure 3.4) [250, 252-254]. This was demonstrated in

a study that showed microparticles that were 0.20 ± 0.04 µm in average diameter resulted

in release of 70% of bone morphogenetic protein 2 (BMP-2) as compared to 12% from

microparticles with an average diameter of 26 ± 6.0 µm after four weeks [250].

Microsphere size and shape can be tuned through various manufacturing techniques with

the most common technique being water-in-oil emulsions induced by mechanical agitation

such as high-speed stirring of a gelatin solution and an organic phase. These emulsions are

then cooled to allow for gelation of the microparticles followed by precipitation [250, 255,

256]. Processing parameters such as mixing speed and solvent selection are used to control

microparticle size; however, this technique typically results in a large particle size

distribution [252, 257]. As an alternative to high-speed stirring, microfluidic devices have

been used to achieve monodispersed particle size distributions. This technique consists of

coaxial flow between an aqueous gelatin solution and an oil-based sheath with each phase

Page 96: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

73

set to different flow rates to control the droplet size [258, 259]. Particles are collected in a

coagulation bath prior to chemical crosslinking. If smaller microparticles are desired, then

electrospraying can be used to form microparticles. This technique applies an electric field

to a low viscosity gelatin solution as it is being extruded from a syringe. Charge repulsion

within the solution droplet at the end of the capillary overcomes the solution surface tension

leading to a solution droplets erupting from the droplet towards a ground or oppositely

charged collector. As the solvent evaporates from the droplet during flight to the collector,

a repulsive threshold is reached within the droplets leading to solution fission into smaller

dried particles [252, 260]. Gelatin microparticles are most commonly crosslinked

following fabrication by chemical reagents (e.g. glutaraldehyde) [261-263]. It can be

difficult to control the crosslinking density and these chemical reagents are typically

cytotoxic [237, 264]. Genipin and carbodiimides are alternative reagents used to provide

greater control over crosslinking of gelatin microparticles with less cytotoxicity [236, 265,

266].

Page 97: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

74

Figure 3.4: Effect of construct surface area-to-volume ratio on growth factor diffusion

from gelatin microparticles. A) Smaller microparticles have shorter diffusion path lengths

leading to rapid release of growth factors; B) larger microparticles have longer diffusion

path lengths and slower release profiles. 2 Scanning electrospun micrographs reprinted

with permission.

The tunable nature of gelatin microparticles makes them suitable for a range of

tissue engineering applications such as angiogenesis [267], cartilage repair [268, 269],

bone regeneration [221], ocular repair [222], and nerve regeneration [266, 270]. Most

notably, their size enables incorporation into larger scaffolds as a method to decouple

growth factor release kinetics from other scaffold design criteria [252, 271, 272]. This was

2 Scanning electron micrographs were reprinted with permission from “Comparison of micro- vs.

nanostructured colloidal gelatin gels for sustained delivery of osteogenic proteins: Bone morphogenetic

protein-2 and alkaline phosphatase”, by Huanan Wang, Otto Boerman, Kemal Sariibrahimoglu, Yubao Li,

John Jansen, and Sander Leeuwenburgh. Biomaterials, 2012, 33(33), 8695-8703. Copyright © 2012

Elsevier Ltd. All rights reserved.

Page 98: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

75

demonstrated in a study that incorporated gelatin microparticles loaded with VEGF into a

porous lithium calcium polyphosphate scaffold for bone repair associated with

glucocorticoids-induced osteonecrosis of the femoral head. Microparticles were fabricated

through emulsion templating and crosslinked in glutaraldehyde prior to diffusional loading

of VEGF [263]. In addition to the use of gelatin microparticles in composite scaffolds,

microparticles can also be directly injected as a slurry of particles for more rapid growth

factor delivery. Hirose et al. reported the delivery of basic fibroblast growth factor (bFGF)

and interferon-beta (IFNβ) from gelatin microparticles as a method to establish a

proliferative vitreoretinopathy disease model. The microparticles were fabricated using

emulsion templating and crosslinked in glutaraldehyde prior to diffusional loading of bFGF

or IFNβ [222]. As a general consideration, direct application of gelatin microparticles may

result in a higher initial burst release that results from immediate exposure to aqueous

solutions as compared to microparticles embedded within a composite.

3.3.2 Gelatin Scaffolds

Several researchers have explored the use of gelatin constructs to act as both a

controlled growth factor delivery vehicle and as a scaffolding material. Gelatin scaffolds

have been fabricated using a variety of methods including electrospinning, microfluidics,

freeze-drying, and porogen leaching. The resulting porous, three-dimensional architectures

template new tissue formation by supporting cell attachment, proliferation, and migration

[45, 53]. These same pores can alter the diffusion path length that affects growth factor

release profiles as previously described in delivery vehicles. Electrospinning has become

Page 99: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

76

one of the most widely used techniques to fabricate gelatin scaffolds that serve as growth

factor matrices. During electrospinning, an electric potential is applied to a gelatin solution

that is constantly flowing from a syringe. Charge repulsion within the solution droplet at

the end of the capillary overcomes the solution surface tension leading to a solution jet

erupting from the droplet towards a ground or oppositely charged collector. As the solvent

evaporates from the solution jet during flight to the collector, nanometer to micron-sized

solid polymer fibers are generated [52]. Electrospun scaffolds are commonly crosslinked

post-fabrication using glutaraldehyde [231] or EDC/NHS [48, 273]. If gelatin-methacrylate

is used, then fibers can be in-situ crosslinked by UV photo-polymerization [230, 239].

Alternatively, reactive electrospinning utilizes a bi-functional crosslinker such as a

diisocyanate to initiate in-situ crosslinking of gelatin fibers during the electrospinning

process [239]. Similar to the microparticles, the electrospinning parameters such as

solvent, distance, and flow rate can be modulated to generate a range of fiber diameters

with larger fibers resulting in longer diffusion path lengths for sustained release profiles

(Figure 3.5) [45, 53, 251]. This was demonstrated in a study that showed electrospun fibers

with an average diameter of 1.0 ± 0.1 µm resulted in 7.7 ng of platelet derived growth

factor (PDGF) as opposed to 4.8 ng from thicker fibers with an average diameter of 3.0 ±

0.2 µm after 20 days [251]. Although electrospinning is the most-widely used fabrication

technique for gelatin scaffolds that serve as growth factor matrices, a variety of fabrication

techniques have been used to produce fibrous gelatin constructs with a range of surface-

area-to-volume ratio and shapes. For example, microfluidic spinning is another common

manufacturing process that is similar to microfluidic microparticle fabrication in that an

Page 100: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

77

aqueous gelatin solution is flowed through an oil-based sheath or in a silicone microchannel

[45, 46]. Differences in flow rates, surface tension, and energy dissipation keeps the two

streams separated. This technique allows for precise control over the architecture and

uniform size of the resultant fibers. Precipitation of the gelatin fibers can also be achieved

through a coagulation bath. Fibers produced by microfluidic spinning range from

nanometers to hundreds of microns [45] and are generally crosslinked by glutaraldehyde

[46], UV photo-initiation [274, 275] following precipitation.

Figure 3.5: Effect of construct surface area-to-volume ratio on growth factor diffusion

from gelatin fibers. The diffusion path length in electrospun constructs are controlled by

fiber diameter with A) thin fibers having shorter diffusion path lengths and rapid release;

B) thick fibers have longer diffusion path lengths and slower release profiles. 3

Representative scanning electron micrographs reprinted with permission from [251].

3 Scanning electrospun micrographs were reprinted with permission from “The effected of controlled

release of PDGF-BB from heparin-conjugated electrospun PCL/gelatin scaffolds on cellular bioactivity and

infiltration”, by Jongman Lee, James Yoo, Anthony Atala, and Sang Jin Lee. Biomaterials, 2012, 33(28),

6709-6720. Copyright © 2012 Elsevier Ltd. All rights reserved.

Page 101: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

78

The breadth of architectures available enables the use of gelatin scaffolds in a range

of applications such as wound healing [231], bone regeneration [230, 276], and

angiogenesis [48, 273]. A primary advantage of fibrous gelatin matrices is that they have

the potential to be applied as a stand-alone treatment for tissue engineering grafts [277].

For example, electrospun gelatin fiber meshes loaded with FGF-2 were fabricated for

potential use as a tissue engineering construct. Gelatin fibers were crosslinked by both EDC

and glutaraldehyde and FGF-2 was bound to gelatin fibers by electrostatic avidin-biotin-

complexes. These composite meshes displayed enhanced cell attachment and proliferation,

key targets for enhanced tissue regeneration [231]. In another application, electrospun

gelatin wraps containing transforming growth factor beta 2 (TGFβ2) were fabricated and

crosslinked by genipin. The scaffolds displayed enhanced proliferation and migration with

potential application as a medial layer of vascular grafts for modulation of the hemostatic

environment [278]. Although fibrous gelatin grafts permit controlled release of growth

factors and support cell proliferation and migration, densely packed fibers can limit cell

infiltration [279, 280].

3.4 FUTURE PERSPECTIVES IN THE MASQUELET TECHNIQUE

Gelatin has evolved as one of the most widely studied growth factor delivery

vehicles due to its native physiochemical properties that enable high loading efficiencies

and tunable crosslinking and fabrication processes that provide a broad range of

mechanisms to sequester growth factors for temporal release during tissue engineering.

Page 102: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

79

This technology meets many design requirements set forth for enhancement of

angiogenesis during management of critical-sized bone defects using the Masquelet

induced membrane technique. However, tissue regeneration is typically regulated by

expression of multiple biochemical cues over various periods. This creates a clear need for

a multifactor release system with independent control over temporal release of each factor.

To this end, we have developed a gelatin-based bimodal release system that can address

this need. This release system consists of two in situ crosslinked gelatin carriers combined

by co-electrospinning.

One of the carriers entail photo-crosslinked gelatin that enables burst release of

growth factors. Functionalization of gelatin with methacrylate groups (gel-MA) enables

gelatin to undertake free radical polymerization in the presence of a photo-initiator and UV

irradiation [28]. In-line loading of the gelatin-methacrylate with a growth factor embeds

the growth factor in the gelatin matrix during polymerization resulting in release that is

governed by swelling-based diffusion. Modulation of the initial functionalization

stoichiometry can tune the crosslink density, and therefore the swelling ratio, to control

release kinetics. This gelatin carrier is useful for delivering angiogenic growth factors that

are potent during the initiation of angiogenesis (e.g. VEGF and ANG-2) [281]. The second

carrier consists of diisocyanate-crosslinked gelatin (gel-NCO) that imparts sustained

growth factor release. A reaction between the amines present on the lysine residues of

gelatin and the diisocyanate crosslinker facilitates polymerization of the gelatin molecules.

The incorporation of a growth factor into the gelatin solution results in conjugation of the

growth factor via a reaction between the diisocyanate crosslinker and its amine groups.

Page 103: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

80

Release of the growth factor requires enzymatic degradation to cleave linkages and allow

for subsequent diffusion as the gelatin matrix swells. Changes to the diisocyanate

crosslinker concentration can tune the crosslink density that regulates the degree of

enzymatic degradation required for release. As such, the gel-NCO carrier is advantageous

for angiogenic factors that have a prevalent role during the latter stages of angiogenesis

(e.g. TGF-β and PDGF) [281]. Co-electrospinning these two gelatin-based carriers

combines distinct material properties and corollary release kinetics to yield a single

construct that can be independently tuned (Figure 3.6). Additional modification of release

kinetics can be achieved by tuning the fibrous properties of the individual release systems

as previously described.

Figure 3.6: Bimodal release of model proteins (FITC-bovine serum albumin and TRITC-

bovine serum albumin) from a single electrospun gelatin-based mesh in collagenase. 4

Figure reprinted with permission from.

4 This figure was reprinted with permission from “Development of a bimodal, in situ crosslinking method

to achieve multifactor release from electrospun gelatin”, by Alysha Kishan, Taneidra Walker, Nick Sears,

Thomas Wilems, and Elizabeth Cosgriff-Hernandez. Journal of Biomedical and Materials Research, Part A,

2018, 106(5), 1155-1164. © 2018 Wiley Periodicals, Inc. All rights reserved.

Page 104: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

81

The bimodal release system shows much promise and could overcome the

limitation of a transient vasculature density in the induced membrane to improve clinical

outcomes. However, the release system requires further investigation to establish clinical

significance. Our previous work has demonstrated tunable release with use of model-

proteins but this potential has yet to be investigated for actual angiogenic factors of interest.

This study is required to elucidate the impact of their isoelectric potentials and molecular

weights on release kinetics. Although crosslinking is presumed to preserve bioactivity of

the growth factor, additional studies are needed to assess the effect of physical and

chemical processing applied during fabrication on bioactivity retention. Finally, in vivo

evaluation of the individual release systems and the bimodal release system is required to

confirm the predicted release kinetics and establish their efficacy in angiogenesis. The

following chapter further investigates the potential of a co-electrospun gelatin-based

bimodal release system as an adjunct therapy for enhancing angiogenesis in the induced

membrane during the Masquelet technique.

Page 105: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

82

Chapter IV: A Multifaceted Matrix to Enhance Angiogenesis and

Provide Infection Control during Bone Regeneration

4.1 INTRODUCTION

Successfully improving the Masquelet technique requires a substrate that provides

antimicrobial prophylaxis and guides formation of the induced membrane for improved

durability and vascularity. Resorbable fibrous meshes offer a platform that can be

engineered to achieve these goals, independent of the PMMA spacer [188, 282-286].

Fibrous meshes produced by electrospinning are of particular interest due to the versatility

in materials and fibrous microarchitecture (e.g. fiber diameter and fiber alignment) that

permits biomimicry of the extracellular matrix for cell scaffolding [54, 287]. Electrospun

meshes are fabricated by applying electrostatic forces to a polymer solution. Modulation

of the electrospinning parameters enable tunable structural properties that can be used as a

template to direct cell behavior and matrix deposition for a more robust membrane with

improved durability [287]. Beyond cell scaffolding, electrospun meshes serve as matrices

for bioactive agents, with tunable release that can be used to further guide cell behavior for

angiogenesis as well as inhibit bacterial growth [288, 289]. To our knowledge, only a few

studies have demonstrated this potential for the Masquelet technique [282, 283].

Yu et al. performed a proof-of-concept study that evaluated the ability of an

electrospsun poly(lactic-co-glycolic) acid (PLGA) wrap to induced formation of a

membrane absent of a PMMA spacer. The electrospun wrap was completely resorbed after

6 weeks. Histological analysis of the membrane revealed a cell-rich layer with expression

Page 106: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

83

of angiogenic and osteogenic growth factors. These results confirmed the ability of the

electrospun PLGA wrap to induce membrane formation. However, no instruction beyond

material cues and scaffolding were provided by the electrospun PLGA to improve

formation of the membrane. The potential of the membrane to promote bone regeneration

with and without autologous bone graft was also assessed. Despite the demonstrated

membrane bioactivity, incomplete bone bridging and poor bone consolidation resulted

from the absence of bone graft and indicated that the PLGA-induced membrane was not

sufficient to promote bone regeneration alone. The lack of comparison between the PLGA-

induced membrane and a PMMA-induced membrane composition further challenged the

proposed potential of the electrospun PLGA-induced membrane. This control is important

to include as to establish the efficacy of wrap as an improved or synergistic solution to the

standard technique [283].

The findings from the initial study provided the fundamentals for the development

of a composite substrate encompassing the electrospun PLGA wrap. In this study, Yu et

al. modified the electrospun PLGA system to contain vancomycin/ceftazidime-loaded

layer and a BMP-2-loaded layer. The potential of the composite substrate was evaluated

on membrane formation absent of a PMMA spacer and subsequent bone regeneration with

bone substitute after 8 weeks. Results demonstrated that the PLGA fibers released the

antibiotics and BMP-2 for up to 4 weeks. The composite substrate also induced formation

of a membrane that was rich in cells, biochemical cues, and blood vessels. Satisfactory

bone bridging and mechanical strength were observed with use of the composite substrate

as compared to treatment without the composite substrate. However, this study similarly

Page 107: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

84

failed to compare these findings to that of a PMMA-induced membrane to confirm

improvement or synergy. Another limitation of this study is that the composite substrate

was not evaluated in an infected femoral defect model to validate efficacy of the

antimicrobial potential [282].

The purpose of this study was to engineer a multifunctional wrap as an adjunct

therapy that can guide membrane formation while inhibiting infection. In preceding

chapters, we have highlighted the ability of a resorbable electrospun PLGA mesh to control

release of antimicrobial agents and demonstrated the antimicrobial potential in treatment

of osteomyelitis. We have also elucidated the ability of a gelatin mesh to control release of

growth factors. Therefore, in this chapter, the capacity of a previously developed photo-

crosslinked gelatin-methacrylate (gel-MA) electrospinning mechanism to enable in-line

loading of vascular endothelial growth factor (VEGF) for enhanced angiogenesis was first

confirmed [290]. Characterization of the in vitro VEGF bioactivity retention was evaluated

to validate the safety of electrospinning and crosslinking on VEGF bioactivity. In vivo

analysis of gel-MA resorption and corollary release of VEGF was assessed in a rat

subcutaneous model to establish proof-of-concept for controlled release. We then utilized

co-electrospinning to combine the VEGF-loaded gel-MA release system with the

gentamicin sulfate-loaded PLGA release system to create the multifunctional wrap. The

co-electrospun wrap was investigated to corroborate dual-fiber populations and

maintenance of wrap integrity after swelling. The retention of the release profiles of VEGF

and gentamicin sulfate from the co-electrospun wrap was then evaluated in comparison to

individually established release profiles. To improve clinical application and prevent

Page 108: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

85

displacement during treatment, the co-electrospun wrap was further modified with tissue

adhesive moieties. In all, these studies provide initial evaluation of an adjunct

multifunctional wrap that can guide membrane formation during the Masquelet technique

with potential to simultaneously inhibit osteomyelitis for improve clinical outcomes.

4.2. MATERIALS AND METHODS

Materials

All chemicals and reagents were purchased from Sigma Aldrich (Milwaukee, WI)

and used as received unless otherwise noted.

Gelatin-methacrylate Synthesis

Gelatin-methacrylate (Gel-MA) was synthesized by dropping 2-isocyanatoethyl

methacrylate (IEMA) into a 5 % (wt/wt) solution of gelatin (porcine, type B) in dimethyl

sulfoxide (DMSO). IEMA was added such that the molar ratio of isocyanate to amine was

4X, with the assumption of 11 lysine residues per gelatin molecule. The reaction was

performed under nitrogen at 37°C with stirring for 3 h. The solution was then dialyzed

against reverse osmosis water for 72 h to remove non-functionalized IEMA and DMSO.

The solution was then frozen and lyophilized to isolate the gel-MA product. The degree

of functionalization with IEMA was quantified using NMR based on the method adapted

from Ovsianikov et al.[291]. Briefly, 10 mg of gel-MA sample was dissolved in 1 mL of

deuterium oxide and analyzed using a 400 MHZ Varian MR-400. The methacrylation ratio

was defined as the percentage of amino groups that were modified in gel-MA. The NMR

Page 109: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

86

spectrum was normalized to the phenylalanine sign (6.9 -7.5 ppm) to account for the

concentration of gelatin. The known ratio of 0.011 mol phenylalanine/100 g gelatin was

considered in the integration of this peak (5 protons) to calculate a ratio corresponding to

approximately 0.05297 mol/100 g [292]. The total amount of primary amine groups in

gelatin is known to be approximately 0.0385 mol/100 g [291]. The two new peaks at 5.4-

5.7 ppm on the gel-MA spectrum corresponding to the methacrylate group were integrated

and used in the following calculation to determine the degree of methacrylation:

𝐷𝑒𝑔𝑟𝑒𝑒 𝑜𝑓 𝑚𝑒𝑡ℎ𝑎𝑐𝑟𝑦𝑙𝑎𝑡𝑖𝑜𝑛 = 𝐼5.7𝑝𝑝𝑚

𝐼7.2𝑝𝑝𝑚 ×

0.05297 𝑚𝑜𝑙

100 ×

100

0.0385 𝑚𝑜𝑙

Electrospinning Gelatin-methacrylate

Gel-MA was dissolved at 7 % (wt/wt) in 2,2,2-trifluoroethanol. Ethylene glycol

dimethacrylate was added as a crosslinker at 20 % (wt/wt) of the polymer and a photo-

initiator, lithium phenyl-2,4,6-trimethylbenzoylphosphinates, was added at 5 wt% of the

polymer. The solution was stirred at 37°C overnight prior to electrospinning.

Pentaerythritol tetrakis(3-mercaptoproprionate) was added at 10 wt% of the polymer

immediately prior to electrospinning to reduce oxygen inhibition and facilitate

crosslinking. The gel-MA solution (1 mL) was then added to a syringe protected from light

and dispensed at 1.0 mL/h (KDS100; KD Scientific; Holliston, MA) through a blunted 18

G needle. The syringe pump was placed 12 cm away from a grounded copper plate and a

voltage source charged to 10 kV (ES30P-5W/DDPM; Gamma Scientific; San Diego, CA)

was used to apply to the needle. A UV lamp was placed above the collector to allow for in

situ photo-crosslinking. Each mesh had a total collection time of 1 h followed by one

Page 110: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

87

additional hour of UV exposure. For VEGF-loaded meshes, 0.2 µg of recombinant human

VEGF (R&D Systems; Minneapolis, MN) reconstituted in a 0.1 % (wt/v) BSA was added

to the electrospinning solution. The loading concentration was selected based on

consideration of the model release kinetics and literature-targeted release concentrations

required to induce formation of a mature vasculature [290, 293].

Water Uptatke

The crosslinking density of the electrospun gel-MA wrap was determined by water

uptake analysis. Electrospun wraps (N=3) were soaked in deionized water (DI) overnight

at 37°C to remove uncrosslinked gelatin and swell the specimens. The wraps were blotted

dry and weighed after immersion to obtain the swollen weight. Specimens were then

vacuum dried overnight and weighed again to obtain the dried weight. Water uptake was

calculated using the following calculation:

𝑊𝑎𝑡𝑒𝑟 𝑢𝑝𝑡𝑎𝑘𝑒 (%) = 𝑤𝑠−𝑤𝑑

𝑤𝑑𝑋 100,

where ws indicates the weight of the swollen specimen and wd is the weight of the dried

specimen.

Endothelial Cell Tubulogenesis

The human umbilical vein endothelial cell line (HUVECs; Promocell; Heidelberg,

Germany) was selected to assess bioactivity retention due to its potential to organize into

tube-like vascular networks [294]. Briefly, HUVECs were first expanded in EGM-2

endothelial cell growth media (Promocell), supplemented with 1% of penicillin-

Page 111: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

88

streptomycin (10,000 U/mL; Thermo Fisher Scientific; Waltham, MA) at 37C. Media was

replaced every 48 h until cells were near 80-90 % confluence.

A specimen (10 x 15 mm2) was cut from an electrospun VEGF-loaded gel-MA

mesh and immersed in EGM-2 endothelial cell growth media, supplemented with 0.2 U/mL

of collagenase up to 7 days, with releasate being collected and replaced with fresh media

at 1, 3, and 5 days. Releasate collected from a blank gel-MA mesh at each time point were

used as the negative control. Releasates and controls were stored at -80°C until further use.

Growth factor-reduced Matrigel (Corning; Corning, NY) was thawed at 4°C overnight. On

the day of use, the Matrigel was transferred onto ice to preserve its liquid state. Matrigel-

coated well plates were prepared by plating 50 μL of Matrigel into a 96-well plate at a

horizontal level that allows the Matrigel to distribute evenly without bubbles. Plates were

incubated for 30 min at 37°C to solidify the Matrigel. Releasates and controls were thawed

at 37°C for 10 min prior to use. HUVECs were reconstituted in releasates or controls at a

density of 40,000 cells/ 100 μL. Cell suspensions were carefully plated on top of the

Matrigel and incubated at 37°C for 4 h. To assess network formation, cells were stained

with Calcein, AM (Invitrogen; Carlsbad, CA) and imaged using a fluorescence microscope

(Nikon Eclipse TE2000-S; Melville, NY). Network formation was captured through raster

patterning of 3 wells/group at each time point. Automated counts of network formation

were achieved using the Angiogenesis Analyzer plugin for ImageJ, which detects

characteristic points and elements of endothelial cell networks [295]. VEGF bioactivity

retention was measured by the ability of released VEGF to encourage morphogenesis of

HUVECs into capillary-like networks as a function of VEGF concentration compared to

Page 112: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

89

the negative controls at each time point. A standard curve of capillary-like networks was

formed based on various concentrations of unprocessed VEGF in cell culture media. This

curve was then used to extrapolate the number of networks that would form based on the

expected released concentrations at each time point that was determined from model

release kinetics. Direct comparison of the network formation in response to unprocessed

VEGF and released VEGF was used to determine the degree of bioactivity retention at each

time point. Bioactivity retention was deemed acceptable at >80 %. Studies were repeated

in triplicate and results are represented in terms of the average networks formed at each

time point for each group.

Animal Care

All procedures were approved by the University of Texas Institutional Animal Care

and Use Committee (IACUC, Animal Use Protocol# 2019-00183).

In Vivo Assessment of Resorption

In vivo resorption rates and corollary release kinetics of VEGF from electrospun

gel-MA meshes were assessed in a pilot study. Eight-week old Sprague-Dawley rats (n =

2/time point) were randomly assigned to blank electrospun gel-MA meshes or electrospun

gel-MA meshes. Animals were anesthetized and placed on a nose cone flowing with 2%

isoflurane (Animal Health International; Greeley, CO) in oxygen at a rate of 2 L/min. The

dorsal surface was clipped and prepped for surgery with povidone-iodine and 70% ethanol.

The dorsum was covered with a sterile drape and a 4-cm incision was made in the central

Page 113: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

90

third of the dorsum. Blunt dissections were made in the left and right dorsolateral areas to

prepare four implant pockets. UV sterilized blank and VEGF-loaded electrospun gel-MA

specimens (1 x 6 cm2, n=2/ rat) were placed in the pockets in a sterile manner. The incision

was then closed with stainless steel surgical wound clips (Braintree Scientific; Braintree,

MA) and all animals were percutaneously administered carprofen (5 mg/kg; Animal Health

International). One week after the initial procedure, all rats were anesthetized and the

wound clips were removed. At 2, 4, and 6 weeks, the rats were euthanized via carbon

dioxide inhalation followed by bilateral thoracotomy to ensure death. The incisions were

opened and any remaining specimens were harvested. To evaluate resorption, harvested

specimens were lyophilized and dried masses at each time point were compared to the

initial mass of the sample. Release of VEGF was assessed using an ELISA kit (R&D

Systems). Briefly, lyophilized specimens were degraded in 200 U/mL of collagenase to

release remaining VEGF loaded in the specimens at each time point. Solutions were

analyzed following the manufactures instructions. In vivo release was confirmed by

comparing the remaining VEGF content at each time to the initial content loaded in

specimens that were not implanted.

Co-electrospinning

Co-electrospun meshes were fabricated by electrospinning the gel-MA with PLGA.

Briefly, a gel-MA solution was prepared as previously described. 50:50 PLGA (acid-

terminated; inherent viscosity range 0.55–0.75 dL/g; DURECT Corp.; Cupertino, CA) was

dissolved in 1,1,1,3,3,3-hexafluoro-2-propanol (HFIP; Halocarbon; Peachtree Corners,

Page 114: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

91

GA) to produce a 40% (w/v) polymer solution. Gel-MA (1 mL) and PLGA (4 mL)

solutions were placed into their respective syringes and were placed on either side of the

mandrel to minimize interaction of the electric fields. Gel-MA electrospinning parameters

were set according to the specifications previously described above. PLGA was

electrospun at a flow rate of 0.7 mL/h through a 20 G blunted needle charged to 8 kV at a

distance of 15 cm from the collector. Fibers were collected at ambient conditions on a

rotating grounded mandrel. The total collection time for gel-MA was 1 h and 4 h for PLGA.

The fabricated meshes were vacuum dried for a minimum of 12 h prior to characterization.

For VEGF-loaded gel-MA fibers, 0.2 µg of rhVEGF reconstituted in a 0.1 % (wt/v) BSA

was added to the electrospinning solution. For gentamicin sulfate-loaded PLGA fibers, 20

mg of gentamicin sulfate powder was dissolved in the electrospinning solution. This dose

was determined by considering the release kinetics obtained from preliminary scouting

studies (data not included) and by considering reported minimum inhibitory concentrations

(MIC; lowest concentration that visibly inhibited bacteria growth) of gentamicin sulfate

for the osteomyelitis-derived strain UAMS-1 [123].

Characterization of Co-electrospun Meshes

Fluorescence microscopy of co-electrospun meshes was performed using a

confocal fluorescence imaging system (MaiTai HP; Spectra Physics; Mountain View, CA).

The gel-MA solution was mixed with 2 wt% fluorescein, and the PLGA solution was mixed

with 0.1 wt% DAPI. These fluorescently-doped solutions were co-electrospun onto a glass

slide attached to the rotating mandrel using the parameters described above.

Page 115: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

92

Co-electrospun meshes were subjected to an aqueous soak to assess delamination

of the fiber populations during swelling. Briefly, 2 x 2 cm2 specimens were cut from co-

electrospun meshes (N=3) and soaked in phosphate buffer saline (PBS). Meshes were

visually inspected for noticeable indications of delamination.

A total of 4 meshes were fabricated for gel-MA, PLGA, and co-electrospun

formulations. One dog-bone specimen was cut from each wrap to yield 4 specimens/group

in accordance with ASTM D1708. Specimens were strained to failure at a rate of 100

%/min based on the initial gauge length using an Instron 3345 uniaxial tensile tester

equipped with a 1000-N load cell and pneumatic side action grips (Instron 2712-019;

Norwood, MA). Stress-strain curves were generated to determine the response of each

electrospun composition under tensile loading.

VEGF Release from Co-electrospun Meshes

VEGF-loaded co-electrospun meshes were fabricated to evaluate release kinetics.

Specimens (1 x 6 cm2, n=3) were placed in 2 mL of PBS at 37°C with shaking. At daily

time points, releasates were removed from each specimen and replaced with fresh PBS.

The amount of VEGF present in the releasate was determined using an ELISA kit. The

percentage of released VEGF was then calculated based on a standard curve and the initial

mass of the VEGF incorporated in the electrospun mesh. Release kinetics of the co-

electrospun mesh was compared to release kinetics of a VEGF-loaded electrospun gel-MA

mesh that was evaluated at the same conditions. Studies were repeated in triplicate and the

Page 116: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

93

data is presented in terms of average cumulative percent release over time and average dose

released (ng).

Gentamicin Sulfate Release from Co-electrospun Meshes

Gentamicin sulfate-loaded co-electrospun meshes were fabricated to evaluate

release kinetics. Specimens (1 x 6 cm2, n=3) were placed in 2 mL of distilled water at 37°C

with shaking. Deionized water was selected, as opposed to phosphate buffer, due to the

low sensitivity of gentamicin sulfate in buffered solution at the low concentrations. At daily

time points, release medium was collected and replaced with fresh deionized water. The

collected release medium was frozen and lyophilized to concentrate the gentamicin sulfate.

The dose released at each time point was determined using a ninhydrin assay. Briefly, 0.5

mL of 2% ninhydrin was added to the lyophilized product and incubated at 120C for 15

min. The ninhydrin solutions were diluted with 1 mL of distilled water to enable

spectrophotometric analysis (Infinite 200 Pro; Tecan; Morrisville, NC) at 570 nm. The

percentage of released gentamicin sulfate was then calculated based on a standard curve

and the initial mass of the gentamicin sulfate incorporated in the electrospun mesh. Release

kinetics of the co-electrospun mesh was compared to release kinetics of a gentamicin

sulfate-loaded electrospun PLGA mesh that was evaluated at the same conditions. Studies

were repeated in triplicate and the data is presented in terms of average cumulative percent

release over time and average dose released (µM).

Dopamine Tissue Adhesive Coating

Page 117: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

94

Dopamine hydrochloride was dissolved at 2 or 20 mg/mL in 10 mM tris. 1 mL of

each solution was then carefully pipetted onto a PLGA electrospun mesh. After 24 h, the

solutions were aspirated and 1 mL of 3 wt% sodium periodate in distilled water was applied

to the meshes. After 1 minute, the sodium periodate solution was aspirated and meshes

were washed in 50 mL of distilled water for 5 h with water changes every hour. Samples

were subsequently frozen and lyophilized prior to characterization. The dopamine-

modified PLGA layer was affirmed using ATR-FTIR spectroscopy (Nicolet iS10;

ThermoFisher Scientific; Waltham, MA) at a resolution of 2 cm-1 for 32 scans.

Adhesion Strength Measurement

Adhesive properties of the dopamine-coated PLGA meshes were determine using

lap-shear tensile stress measurements in accordance with the ASTM standard F2255-05,

with some modification. To assess adhesion to bone, fresh porcine cortical bone with in-

tact periosteum was cut into 2.5 x 5 cm2 segments with uniform thickness and hydrated in

PBS for immediate use. Un-modified and adhesive PLGA meshes (N=4) were fabricated

and stored in a dry location at room temperature. A 2.5 × 1 cm2 specimen was removed

from each mesh to account for batch variability. The porcine test substrates were removed

from PBS and blotted with sterile gauze to remove excess PBS prior to coming into contact

with the un-modified PLGA specimens or adhesive PLGA specimens. The specimens were

positioned between two tissue substrates with a 1 cm overlap. They were then compressed

with a force of approximately 1 N to allow for the bond to set. Specimens were further

conditioned in a humidity chamber at 37°C for 30 min prior to being strained to failure at

Page 118: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

95

a rate of 5 mm/min using an Instron 3345 uniaxial tensile tester equipped with a 100-N

load cell and pneumatic side action grips (Instron 2712-019). The maximum strength and

failure strain were recorded. The adhesive strength was calculated by maximum strength

divided by the initial bond area.

Statistical Analysis

Data averages are accompanied with ± standard deviation. Statistical analysis was

performed utilizing a standard one-way ANOVA with Tukey’s post-hoc analysis unless

otherwise indicated in figure captions. Statistical significance was accepted at P<0.05.

4.3 RESULTS

Fabrication and Characterization of Gelatin-methacrylate Wrap

Controlled delivery of angiogenic factors has been demonstrated to improve

angiogenesis in tissue engineering applications. Implementation of this technology can

address limitations of the Masquelet technique for treatment of critical-sized bone defects.

In this present study, we used a previously developed in situ crosslinking mechanism to

enable in-line loading and encapsulation of VEGF during gel-MA electrospinning [290].

NMR analysis confirmed that approximately 85% of gelatin lysines were functionalized

with methacrylate groups (Figure 4.1). Assessment of the crosslinking density confirmed

a water uptake of 282 ± 80 % indicating successful photo-polymerization of the gel-MA

wrap.

Page 119: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

96

Figure 4.1: NMR spectra of A) gelatin and B) gel-MA used to quantify functionalization.

To confirm VEGF encapsulation and evaluate the effect of fabrication on

bioactivity retention, HUVECs were suspended in VEGF releasate collected at each time

point and seeded onto Matrigel®. Success was determined by the ability of HUVECs to

organize into significantly greater numbers of capillary-like networks as a function of

VEGF concentration as compared to the blank gel-MA releasate (negative control). A

standard curve of network formation was created to confirm cellular response to

unprocessed VEGF at increasing concentrations (Figure 4.2). VEGF-loaded gel-MA

releasate resulted in 27 ± 6 networks after 1 day of release, 14 ± 6 networks after 3 days of

release, and 11 ± 5 networks after 5 days of release. The quantity of networks formed at

each time point was significantly greater than the quantity formed by blank releasate at the

Page 120: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

97

corresponding time point. These results indicated that VEGF bioactivity was not severely

impeded by fabrication (Figure 4.3). The degree of bioactivity retention was determined

at each time point by comparing the number of releasate-induced networks to the number

of networks that formed in response to unprocessed VEGF at theoretical released

concentrations. The theoretical concentration at each time point was derived from

previously determined release kinetics using model growth factors [290]. Results specified

that the estimated bioactivity retention was greater than 80% by the fifth day of release,

signifying that the in situ crosslinking mechanism successfully encapsulated VEGF and

that the bioactivity was retained following fabrication.

Page 121: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

98

Figure 4.2: Capillary-like network formation in response to unprocessed VEGF. A)

Representative images of network formation induced by increasing concentrations of

unprocessed VEGF. Cells stained with calcein-AM. Scale bar is 200 µm. B) Quantified

network formation/field of view corresponding to the representative images.

Figure 4.3: Evaluation of the bioactivity of released VEGF from electrospun gel-MA

meshes. Representative images show capillary-like network formation corresponding to

blank releasate and releasate from VEGF-loaded meshes. Cells stained with calcein-AM.

Scale bar is 200 µm. Graph displayed quantifies the network formation over 5 days of

VEGF release. * indicates statistical differences with respect to the network formation

induced by blank releasate at each time point.

After evaluating the effect of fabrication on loading and bioactivity retention, initial

in vivo studies were conducted to evaluate gel-MA resorption and corollary release. VEGF-

loaded and blank gel-MA specimens were implanted subcutaneously into the dorsum of

Sprague-Dawley rats and resorption and release were assessed at 2, 4, and 6 weeks. Studies

Page 122: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

99

indicated that gel-MA wraps were present at 6 weeks; though, there was over 40% mass

loss as compared to non-implanted specimens, suggesting that gel-MA resorption occurred

(Figure 4.4A). Analysis of the in vivo release kinetics at each time point with an ELISA

assay revealed that the electrospun gel-MA wrap released its entire VEGF load by 6 weeks,

despite over half of the gel-MA mass remaining at this time (Figure 4.4B). These findings

confirmed that VEGF release from the electrospun gel-MA wrap is primarily governed by

swelling-mediated diffusion rather than resorption, as total release was completed before

resorption. In all, these initial in vivo studies provide proof-of concept of controlled release

of VEGF from the electrospun gel-MA wrap for angiogenesis.

Figure 4.4: In vivo evaluation of VEGF release kinetics from electrospun gel-MA in a rat

subcutaneous model. A) Mass loss of blank and VEGF-loaded gelatin-methacrylate

meshes over 6 weeks. B) Corresponding release of VEGF from gelatin-methacrylate

meshes.

Development of a Multifunctional Wrap

Page 123: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

100

Treatment of critical-sized bone defects with the Masquelet technique is limited by

induced membrane bioactivity and durability. Bacteria colonization of implants further

complicates treatment with high incidences of osteomyelitis. We used co-electrospinning

to combine the VEGF-loaded gel-MA release system and a gentamicin-loaded PLGA

release system into a multifunctional wrap with the potential to enhance the induced

membrane and reduce infection as an adjunct therapy. It was hypothesized that combining

the two release systems would allow independent control over multifactor release. Dual-

fiber population was confirmed via fluorescent labeling of each solution with fluorescein

(gel-MA) and DAPI (PLGA) (Figure 4.5).

Figure 4.5: Schematic of co-electrospinning apparatus. Blow out image depicts dual-

fiber population. Fluorescein (green) fibers are gel-MA and DAPI (blue) fibers are

PLGA.

Page 124: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

101

The co-electrospun wrap was assessed for stability and strength via aqueous

immersion and tensile properties. Delamination was not observed under swelling

conditions confirming stability of the co-electrospun wrap. Furthermore, the co-

electrospun wrap exhibited greater tensile strain and strength than the electrospun gel-MA

wrap alone (Figure 4.6). These results suggested that the co-electrospinning gel-MA with

PLGA improves requisite mechanical properties of the wrap.

Figure 4.6: Stress-strain response for each electrospun wrap under tensile loading. Blue

line indicates electrospun gel-MA, red line indicates electrospun PLGA, and green line

indicates co-electrospun gel-MA and PLGA. Arrows denotes tensile failure at the

associated strain.

Page 125: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

102

VEGF and gentamicin sulfate release from the co-electrospun wrap were evaluated

daily over 14 days to validate retention of the respective release kinetics when combined.

VEGF release kinetics from the co-electrospun wrap was assessed with an ELISA assay

and was shown to have a burst release comparable to the gel-MA wrap. Similarly, there

was no significant difference between the cumulative release after 14 days (97.8 ± 2.5 %

for gel-MA wrap and 86.1 ± 3.1 % for co-electrospun wrap) indicating that co-

electrospinning did not impact release kinetics of the electrospun gel-MA component

(Figure 4.7A). Analysis of the dose of VEGF that was released daily from the co-

electrospun wrap revealed an overall decrease in the released amount at each time point as

compared to the gel-MA wrap (Figure 4.7B). This decrease was attributed to off-target

fiber collection in the electrospinning set-up due to the build-up of fibers on the

surrounding electrospinning apparatuses. The loss of fiber collection resulted in daily dose

of VEGF below the targeted concentration range for angiogenesis (0.5 -11 ng/ml) over 10

days [293, 296]. A similar trend was observed for the comparison of release kinetics of

gentamicin sulfate from the co-electrospun wrap and the PLGA wrap over 14 days (Figure

4.8). However, the lower dose that was released from the co-electrospun wrap at each time

point was still greater than the hypothesized MIC concentration for treating osteomyelitis

(1µM) [123]. Overall, this data demonstrated that co-electrospinning enables independent

control over multifactor release from a single adjunct therapy.

Page 126: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

103

Figure 4.7: In vitro release of VEGF in PBS over 14 days. A) Cumulative release of

VEGF from electrospun gelatin-MA as compared to release from the co-electrospun

wrap. B) Corresponding daily release of VEGF from gel-MA as compared to the co-

electrospun wrap. Red-dashed line indicates the lowest-targeted VEGF concentration.

Figure 4.8: In vitro release of gentamicin sulfate in water over 14 days. A) Cumulative

release of gentamicin sulfate from electrospun PLGA as compared to release from the co-

electrospun wrap. B) Corresponding daily release of gentamicin sulfate from PLGA as

compared to the co-electrospun wrap. Red-dashed line indicates the h ypothesized MIC.

Tissue Adhesive Multifunctional Wrap

Previous studies have established the potential of dopamine to provide tissue

adhesive properties to material substrates. Electrospun PLGA meshes were coated with

Page 127: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

104

dopamine using the reported dip coating method to improve clinical application and

prevent displacement during healing (Figure 4.9A). Infrared spectral analysis confirmed

the presence of the dopamine coating indicated by dopamine specific peaks on the PLGA

wrap (Figure 4.9B). Lap shear analysis was used to determine the tissue adhesive potential

of the dopamine-coated PLGA mesh as compared to an un-modified PLGA mesh.

Specimens were applied to fresh porcine cortical bone with an in-tact periosteum and

evaluated under tensile loading. The average shear strength of the dopamine-modified

PLGA mesh increased with higher dopamine concentrations (Figure 4.10C). The average

shear strength of the un-modified PLGA mesh was not included due to shear forces being

below the detection limit of the load cell. Overall, these result demonstrated the potential

of dopamine to confer tissue adhesive properties to biomaterials.

Page 128: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

105

Figure 4.9: Fabrication of dopamine-modified PLGA wrap for tissue adhesion. A)

Schematic of the dopamine dip-coating process. B) ATR-FTIR of the un-modified PLGA

mesh (light gray line), pure dopamine (dark gray line), and dopamine-modified PLGA

mesh (black line).

Page 129: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

106

Figure 4.10: Evaluation of a dopamine-modified wrap on tissue adhesion. A) Schematic

of proposed reaction with the periosteum. B) Representative image of the set-up for lap

shear testing the dopamine-modified PLGA mesh with bone. C) Average maximum shear

strength of the dopamine-modified PLGA mesh coated with increasing concentrations of

dopamine. Results of the un-modified PLGA mesh were not included in the graph due to

the shear forces being below the detection limit of the instrument. * indicated statistical

differences with respect to the 2 mg/mL coating concentration.

4.4 DISCUSSION

The Masquelet induced membrane technique is commonly used for management of

critical-sized bone defects. Despite the potential of the induced membrane to aid bone

regeneration, the durability of it varies depending on the topography of the spacer and the

Page 130: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

107

anatomical location. This limits surgical handling and can have adverse effects on cellular

responses. Another limitation is vascular degeneration which reduces the bioactivity of the

induced membrane [15-17]. This variability in membrane bioactivity leads to unpredictable

clinical outcomes [6, 9, 26]. Apart from the bioactive potential of the induced membrane,

osteomyelitis further complicates clinical outcomes [6, 297]. Osteomyelitis can cause

tissue necrosis and require a revision surgery if the infection is not adequately addressed

[6]. Local delivery from antibiotic-loaded PMMA spacers are clinically used as an

alternative to overcome limitations of systemic delivery. However, PMMA carriers are

limited by suboptimal release kinetics [297]. To date, there does not exist a commercial

product that has the complexity to concurrently address the constraints associated with the

Masquelet technique. Effectively improving the technique will require the use of an adjunct

substrate that simultaneously guides formation of the membrane while preventing

infection.

Accordingly, we aimed to develop a resorbable multifunctional wrap using

biomaterial scaffolding and drug delivery that directs cellular responses, enables tissue

remodeling, and inhibits bacterial growth. It is well known that the extracellular matrix has

a significant role in governing the mechanical properties of the induced membrane [11,

54]. Therefore, the resorbable wrap was fabricated by electrospinning to generate a

template to guide cellular adhesion and matrix deposition and well as to guide cellular

behavior for improved durability. Another advantage of electrospinning is the ability to co-

electrospin. This technique generates multipolymer fibrous meshes with disparate material

properties that can be tuned independently of each other [54]. Co-electrospinning was used

Page 131: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

108

to combine gelatin and PLGA into a multifaceted wrap. Gelatin-based fibers are of

particular interest due to both the matrix-like fibrous structure and the RGD (arginine-

glycine-aspartate) ligands that inherently promote cellular adhesion [298]. Gelatin also has

potential as a carrier for VEGF, a potent regulator of angiogenesis, due to its mild

processing conditions [299]. Angiogenesis is a process by which new blood vessels form

from pre-existing blood vessels. Biochemical cues, such as VEGF, stimulate endothelial

cells and smooth muscle cells that are organized into a pre-existing blood vessel to

destabilize, migrate, proliferate, and organize into vascular tubes. These newly formed

tubes are able to form a stable anastomosis with the pre-existing vascular network leading

to a greater vascular network [300]. Controlled delivery of VEGF between 0.5 to 11 ng/mL

daily over 10 days has been demonstrated to increase mature and stable (i.e. persisting

independently of VEGF delivery) capillary formation by approximately 50% [293, 296].

The release of VEGF from a gelatin wrap within this therapeutic range could enhance

angiogenesis to increase the vasculature and corollary bioactivity of the induced

membrane. In the present study, we have used photo-crosslinked gel-MA to enable

controlled resorption and release of VEGF. Modulation of the gel-MA crosslink density

can be used to tune swelling-based diffusion of embedded VEGF [290]. Furthermore,

PLGA was selected as the additional electrospun polymer due to the demonstrated capacity

to control release of gentamicin sulfate. Gentamicin sulfate is a broad-spectrum antibiotic

that inhibits protein synthesis by targeting bacterial ribosomes. Controlled delivery of

gentamicin sulfate from PLGA above the MIC (1 µM) can potentially inhibit viability of

osteomyelitis-derived isolates like methicillin-susceptible Staphylococcus aureus (MSSA)

Page 132: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

109

[123]. Modification of the chemical and fibrous properties of PLGA offer many

mechanisms to tune diffusion of gentamicin sulfate [46, 47]. In addition, these properties

can be tuned to control the mechanical properties and impart additional strength to the

multifunctional wrap.

To enhance angiogenesis, the electrospun gel-MA was loaded with VEGF based on

consideration of target release concentrations and model release kinetics identified in

previous studies [290, 293]. It is well known that growth factors are sensitive to

environmental conditions; therefore, we first evaluated the effects of fabrication and post-

processing on VEGF bioactivity using an endothelial cell tubulogenesis assay, whereby

endothelial cells organize into capillary-like networks in response to VEGF concentration.

This study confirmed that electrospinning did not severely reduce the bioactivity of VEGF

loaded into electrospun gel-MA wrap. These results are in-line with past studies that

demonstrated significant bioactivity retention of VEGF following electrospinning [301,

302]. In vivo evaluation of the VEGF-loaded gel-MA wrap in a rat subcutaneous model

showed that VEGF released over a month and was exhausted before complete resorption

of the wrap. This was expected as previous studies have revealed that the release kinetics

of proteins from gel-MA are primarily governed by swelling-based diffusion rather than

degradation-based diffusion [237, 290]. However, the overall rate of release and resorption

is slower than what has been modeled in previous in vitro studies in our laboratory [290].

This was not unforeseen, as modeling the in vivo environment has proven to be challenging

and can only be used to surmise resorption and release [55]. Slower release and resorption

rates may be of advantage due to the potential to benefit the overall goal of sustaining

Page 133: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

110

release of VEGF. Ultimately, the findings from these studies serve as initial proof-of-

concept for a VEGF-loaded electrospun gel-MA wrap capable of enhancing angiogenesis.

Dual-collection of the gel-MA and PLGA fibers were confirmed via fluorescent

imaging. The requisite tensile properties of the co-electrospun wrap were attributed to the

PLGA due to the higher tensile strain and strength of the PLGA wrap as compared to the

gel-MA wrap. The strength was also greater than what has been reported for the induced

membrane when under tensile stress [32]. This suggests that integration of the co-

electrospun wrap into the induced membrane can impart additional structural support

during formation of the membrane. Furthermore, the release kinetics of the co-electrospun

wrap for VEGF and gentamicin sulfate were evaluated. Both fiber populations in the co-

electrospun wrap retained relatively similar release profiles as compared to the individual

release systems. These results confirmed the ability of co-electrospinning to enable

bimodal release with independently tuned release kinetics. However, the concentration

released from each fiber population in the co-electrospun wrap was lower than what was

exhibited for the individual wraps. This was attributed to off-target collection of the fibers

due to the charge repulsion of the two electrospinning jets [303]. Reduced collection of the

fibers lowers the amount of drug available for release. Although this is could be

problematic for angiogenesis, released doses of gentamicin sulfate were still above the

targeted concentration for antimicrobial activity. These results indicated that VEGF-loaded

gel-MA will need to be electrospun for a longer period to compensate for the reduction of

VEGF available due to off-target collection.

Page 134: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

111

To improve application of the co-electrospun wrap, a dopamine hydrochloride

(dopamine) coating was applied via dip-coating [304]. Dopamine has been identified as a

derivative of the foot protein that allows aquatic mussels to adhere to inorganic and organic

wet surfaces under high shear stress due to water flow using both covalent and non-

covalent interactions. This discovery has led to the incorporation of dopamine in

biomaterials to promote tissue adhesion. Oxidation of the dopamine catechol enables

covalent bonding with primary amines by nucleophilic attack [304, 305]. We hypothesized

that coating the co-electrospun wrap with dopamine would facilitate attachment to the

periosteum of the host bone and prevent the need of suturing to secure the wrap, as suturing

imparts further tissue damage. Another advantage of incorporating dopamine into the co-

electrospun wrap is the strong affinity of cells to dopamine coatings [306, 307]. Ku et al.

demonstrated significantly enhanced HUVEC adhesion, viability, and stress fiber

formation on dopamine-coated polycaprolactone nanofibers as compared to un-modified

nanofibers and gelatin-coated nanofibers [306]. Similarly, Lee et al. reported on the

potential for polydopamine-coated substrates to support fibroblast adhesion [304].

Polydopamine coatings have been shown to have higher adsorption of serum proteins like

fibronectin as compared to unmodified surfaces. It has been hypothesized that cell adhesion

to dopamine coatings is governed by increased cellular affinity to the adsorbed serum

proteins [258]. Therefore, incorporation of dopamine can exert additional cell binding

properties to improve the composition and durability of the induced membrane. In vitro

analysis of the dopamine-modified PLGA demonstrated the potential for tissue adhesion

of the co-electropsun wrap to the periosteum as compared to un-modified PLGA. These

Page 135: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

112

results suggested that a dopamine coating can be implemented to effectively secure the

wrap around the defect without suturing. In vitro studies to evaluate the effect of dopamine

on cell adhesion are currently under investigation.

In all, the co-electrospun wrap offers bimodal release of multiple factors to enhance

angiogenesis and prevent infection. The combination of this technology with biomimicry

of the native matrix structure could be used to guide formation of the induced membrane

to standardize clinical outcomes following the Masquelet technique. The current studies

presented here establish initial evaluation of the multifunctional electrospun wrap with

sustained bioactivity, optimize handling, controlled release, and tissue adhesiveness.

However, there were limitations of these studies that require further investigation. In vivo

angiogenesis in response to the electrospun VEGF-loaded gel-MA mesh has yet to be

investigated to confirm efficacy of the treatment. These studies would need to evaluate the

change in vasculature density and CD31+ blood vessels, which are specific to mature blood

vessel, following treatment as compared to no treatment and blank meshes. Preliminary

studies are in progress to confirm the potential of released gentamicin sulfate to reduce

infection in an osteomyelitis model and improve bone regeneration as compared to no

treatment and a PMMA bone cement carrier. Fibrous properties can be altered to control

release kinetics if bacterial density is above the critical threshold known to cause infection

[308]. Selection of a different PLGA co-polymer ratio can also modulate release. Another

proposed advantage of the multifunctional electrospun wrap is the ability to improve the

durability of the induced membrane for better surgical handling by providing templating

and biochemical signaling. Cellular behavior in response to the co-electrospun wrap was

Page 136: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

113

not experimentally investigated in the present study but cellular adhesion and proliferation

in the presence of gelatin and dopamine is established in the literature [298, 306].

Following completion of the aforementioned studies, in vivo assessment of the

multifunctional electrospun wrap with the Masquelet induced membrane technique will be

required to determine its efficacy in guiding formation of the membrane for improved

healing outcomes.

4.5 CONCLUSION

The aim of this study was to engineer a device that can be implemented to reduce

variable clinical outcomes achieved by the Masquelet induced membrane technique. The

studies performed have established initial evaluation of an adjunct multifunctional

electrospun wrap that can influence formation of the induce membrane rather than relying

on the non-guided biological process. This adjunct wrap has been designed to improve

durability for improved surgical handling, enhance angiogenesis to increase the vasculature

and corollary bioactivity, and provide infection control. The bioactivity retention of VEGF

was confirmed following in-line loading into electrospun gel-MA. In situ crosslinking of

gel-MA exhibited sustained in vivo release of VEGF. Co-electrospinning enabled the

combination of the angiogenic release system with the antibiotic release system. The co-

electrospun wrap possessed enhanced mechanical properties with the capacity for bimodal

release of VEGF and gentamicin sulfate. Bimodal released profiles from the co-electrospun

wrap validated the potential to independently control angiogenesis and provide infection

control from a single substrate. Additionally, the ability exert tissue adhesive properties

Page 137: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

114

was evaluated and confirmed the potential improve surgical application of the adjunct.

Overall, this work defines the fundamentals for an engineered substrate that can guide

formation of the induced membrane to improve treatment of critical-sized bone defects.

Future studies will build on these findings by further characterizing the multifunctional

electrospun wrap in in vitro cell culture and in relevant animal models.

Page 138: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

115

Chapter V: Conclusion

5.1 SUMMARY

The induced membrane has a critical role in facilitating bone regeneration during

the Masquelet technique [9]. However, unpredictable clinical outcomes highlight the need

for a method to better guide formation of the induced membrane. Several research attempts

have been made to modify the spacer for improved formation of the induced membrane,

with the majority of these attempts focused on altering expression of biochemical cues,

enhancing angiogenesis, and increasing durability of the membrane [31-36]. However, no

single approach has definitively guided formation better than the standard technique. An

ideal approach would augment the Masquelet technique with a platform that offers cellular

recruitment, guides cell behavior, and serves as a template to direct matrix deposition and

remodeling for improved durability and enhanced angiogenesis. Due to the frequent

incidence of osteomyelitis associated with critical-sized defects, the approach should also

encompass infection control [6, 103, 104]. The platforms discussed in this work provide

the framework for the development of a multifaceted adjunct wrap with potential to address

these needs.

We first compared the efficacy of two distinct antimicrobials released from a

resorbable fibrous mesh in the treatment of osteomyelitis. The resorbable mesh was

electrospun to allow direct encapsulation of gentamicin sulfate or gallium maltolate into

the PLGA fibers. Release kinetics were then evaluated to confirm sustained release above

Page 139: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

116

the hypothesized MIC for each antimicrobial [123, 141]. Not only was sustained release

above the hypothesized MICs achieved, but it lasted longer than the critical period when

bacteria is susceptible to antimicrobials (72 h) [207, 208]. Next, antimicrobial meshes were

cultured with an osteomyelitis-derived isolate to determine the potential of released drug

to inhibit growth. The sub-optimal antimicrobial activity of gallium maltolate led to further

evaluation of the MIC and MBC for each antimicrobial. The poor antimicrobial activity of

the gallium-loaded PLGA mesh was attributed to a higher calculated MIC as compared to

the hypothesized MIC. The antimicrobial activity of each drug was then evaluated against

its cytotoxicity on osteoblasts to establish the safety and efficacy. From these studies, the

gentamicin sulfate-loaded PLGA mesh was selected as a candidate adjunct wrap to treat

osteomyelitis during the Masquelet technique.

We then developed a resorbable wrap to enhance formation of the induced

membrane by electrospinning a photo-crosslinked gelatin-methacrylate (gel-MA) mesh

loaded with VEGF. The resorbable fibers serve as scaffolding to support cell attachment

and guide cell behavior and matrix deposition during tissue remodeling for improved

durability. Delivery of VEGF from the gel-MA mesh can provide further instruction on

cellular behavior for enhanced angiogenesis during formation. VEGF bioactivity retention

following fabrication and in vivo release were evaluated as an initial proof-of-concept study

for controlled growth factor delivery. Electrospinning did not impede VEGF bioactivity

and enabled controlled release up to 4 weeks in vivo. To engineer a substrate that provided

infection control and enhanced formation of the induced membrane, we adapted an

electrospun gelatin bimodal release system developed by our lab [290]. This modified

Page 140: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

117

bimodal release system was fabricated by co-electrospinning the gentamicin sulfate-loaded

PLGA platform selected in the previous section with the VEGF-loaded photo-crosslinked

gel-MA platform. The combination of the two platforms not only resulted in bimodal

release of VEGF and gentamicin sulfate with the potential to be independently tuned, but

also resulted in a more structurally stable wrap that can improve surgical application and

further guide cellular behavior.

In summary, successfully improving the Masquelet technique requires infection

control and enhancement of the induced membrane. This work underscores the potential

of this multifaceted substrate to serve as an adjunct wrap to enhance formation of the

induced membrane during the Masquelet technique for improve bone regeneration through

1) antimicrobial activity, 2) support for cellular attachment and guided matrix deposition,

and 3) instruction on cellular behavior to promote angiogenesis.

5.2 SIGNIFICANCE OF WORK

By combining biomaterial scaffolding with drug delivery and biochemical

signaling, an adjunct substrate has been engineered to addresses several limitations of the

Masquelet technique. Herein, we describe novel mechanisms imparted into this substrate

to achieve these goals and the impact that the discoveries from this work will have in the

broader field.

In the first section, two antimicrobial wraps were investigated to establish their

potential as treatments in osteomyelitis. The findings from these studies contributed to the

growing area of research containing few reports on the potential of electrospun

Page 141: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

118

antimicrobial wraps as adjunct therapies in bone regeneration. Studies that have evaluated

electrospun antimicrobial wraps as adjunct therapies in infected bone defects did not

evaluate them using relevant osteomyelitis-derived isolates [169, 188]. Our work has

provided analysis of electrospun antimicrobial wraps against an osteomyelitis-derived

isolate to establish better clinical relevance. Furthermore, existing electrospun

antimicrobial wraps used in treatment of osteomyelitis often contain antibiotics [169, 188].

This work highlighted the potential of electrospinning to enable incorporation and

controlled release of a novel antimicrobial, gallium maltolate. Gallium maltolate has been

investigated in treatment for chronic wounds [141, 212]; however, this is the first study to

evaluate its potential in treatment for osteomyelitis. We provided the first known report of

the MIC and MBC for gallium maltolate against an osteomyelitis-derived isolate. These

results build on the fundamentals of gallium maltolate as a broad-spectrum antimicrobial

agent. The MIC of gentamicin sulfate has been discussed in the treatment of osteomyelitis

but not the MBC [123]. Our work has identified the MBC of gentamicin sulfate which can

be used to guide future development of carriers for this drug. Moreover, very few studies

have investigated the cytotoxicity of antimicrobials released from electrospun wraps on

osteogenic cell lines. Those that have seldomly compare the cytotoxicity of the drug to its

antimicrobial activity as to determine if the antimicrobial will be released at concentrations

that are both safe and effective (selectivity index) [169, 188]. Our studies identified the

selectivity indices for each antimicrobial and determined that gentamicin sulfate was a

safer and more effective drug than gallium maltolate in the treatment of osteomyelitis.

Page 142: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

119

These findings further expand the knowledge of antimicrobials available to effectively treat

osteomyelitis.

The candidate antimicrobial wrap was then combined with an electrospun bioactive

wrap to generate a multifaceted therapy. To our knowledge, this work established the first

resorbable adjunct wrap to guide membrane formation with cellular recruitment,

templating, and biochemical signaling. An electrospun photo-crosslinked gel-MA mesh

that was previously developed in our lab was utilized to provide scaffolding and

instructions for cells through biomimicry of the native matrix fibrous structure and

bioactivity of the RGD ligands on gelatin [290]. These features offer a framework to

actively guide cellular behavior during membrane formation as compared to other

researched substrates that only rely on scaffolding. In addition, the gel-MA mesh was

loaded with VEGF to promote angiogenesis. Previous work has established the potential

of this gel-MA mesh to provide controlled release of model-proteins for bone regeneration

[290]. We demonstrated significant in vitro bioactivity retention as well as in vivo release

of VEGF from the gel-MA mesh up to 4 weeks. These results further contribute to the

development of this gelatin release system for controlled delivery of growth factors. These

findings also build upon the sparse reports that describe bioactivity retention of growth

factors following in-line loading into electrospun fibers. By co-electrospinning the VEGF-

loaded gel-MA mesh with the gentamicin sulfate-loaded PLGA, we achieved bimodal

release from a single construct with the potential to be independently tuned. Co-

electrospinning also enables multiple material properties to be combined. This technique

could provide additional mechanical cues that further guide cell behavior during membrane

Page 143: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

120

formation. The increased strength of the wrap imparted by the PLGA fibers can also be

used to reinforce the mechanical properties of induced membrane during formation until

complete remodeling is achieved. Ultimately, these methods enable the wrap to address

limitations of osteomyelitis and the induced membrane simultaneously.

The individual electrospun platforms that were combined to overcome the

limitations of the Masquelet technique can also be used to address other indications in bone

healing. For example, compound fractures are often exposed to contaminants that can lead

to osteomyelitis and delay or prevent healing. The gentamicin sulfate-loaded PLGA wrap

can be used as a stand-alone local prophylaxis to overcome this drawback. Although the

risk of infection is lower in non-compound fractures, there can still be significant damage

to the bone resulting in critical-sized defects that require bone grafting. Allografts are

prevalently used in critical-sized defects that require a substantial amount of graft and

structural support due to their availability as compared to autografts. However, sterilization

and processing of allografts removes the periosteum, which contains cellular and vascular

constituents that confer bioactivity. This results in poor bone union and mechanical

strength [309, 310]. Allograft revitalization has since become a major objective in bone

regeneration. The electrospun gel-MA wrap can serve as a bioactive carrier for growth

factors to emulate the biochemical signaling that is present during periosteum-mediated

healing and guide cell behavior to enhance regeneration. This technology can also augment

synthetic bone grafts that have superior biocompatibility, mechanical properties, and

tunable degradation rates but that lack bioactivity to actively promote healing. When

contamination and significant bone loss are of concern in severe compound fractures, the

Page 144: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

121

multifunctional electrospun wrap can augment bone grafts to address both indications

simultaneously. In all, the ability to address multiple limitations places this work at the

forefront of innovation in bone regeneration.

We have developed a multifunctional adjunct wrap capable of simultaneously

inhibiting bacterial growth and guiding formation of the induced membrane to address

these needs. The overall significance of this work includes expanding knowledge of

antimicrobials available to effectively treat osteomyelitis, imparting structural and

biochemical cues for active guidance of cellular behavior and tissue remodeling, and

combining multiple release systems to achieve bimodal release from a single construct.

Although the mechanisms described here were aimed at improving the Masquelet

technique, the broader impact lies within the development of independent electrospun

platforms that be used in a variety of bone regeneration applications.

5.3 CHALLENGES AND FUTURE PERSPECTIVE

The work presented here describes several advances in improving the Masquelet

technique. However, further investigation must be performed for clinical translation.

A primary focus of this work has been on evaluating and identifying an adjunct

antimicrobial therapy in the treatment of osteomyelitis. Although we have demonstrated

the potential of the gentamicin sulfate-loaded PLGA wrap to reduce bacterial

contamination at released concentrations that are safe on osteoblasts, bone healing is

orchestrated by more than just osteoblasts. Additional work is needed to evaluate

cytotoxicity of gentamicin sulfate on other cell lines such as chondrocytes and endothelial

Page 145: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

122

cells to better determine the potential efficacy of the wrap. It is also important to evaluate

the high burst release from the wrap on nephrotoxicity due high incidences of gentamicin

sulfate-induced nephrotoxicity [311]. The antimicrobial wrap is electrospun for 4 h to

achieve a relatively high drug load that enables in vitro detection of release kinetics during

the sustained release phase. This results in concentrations released that are significantly

greater than the calculated MIC and MBC of gentamicin sulfate. However, no adverse

effects are expected due to the total load of gentamicin sulfate in the PLGA wrap being

below (20-fold decrease) the clinically-administered dose known to induce necrosis and

renal dysfunction in animals [311]. The most robust method to evaluate nephrotoxicity will

be in vivo analysis due to the challenges associated with modeling cellular responses in

vitro. If released doses are found to be nephrotoxic, then a reduction of the wrap’s thickness

will be required to reduce the available drug load and subsequent released concentrations.

The evaluation of in vivo antimicrobial activity is also warranted in an infected bone defect

model. It has been reported that there is a bacterial threshold (>103 CFU/g of tissue) for

reliable infection that impedes bone healing [308]. Current studies are in progress to

evaluate the remaining bacterial load and bone healing following treatment with the

gentamicin sulfate-loaded PLGA wrap. These studies are important to validate in vitro

modeling of antimicrobial activity and cytotoxicity. They will also establish the potential

efficacy of local delivery at relatively low doses over a shorter duration as compared to

systemic treatment and other local delivery devices. If the released concentrations are

found not to be effective, then additional modification of the wrap will be necessary. One

of the advantages of this electrospun wrap is the ability to be adapted as needed to achieve

Page 146: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

123

clinical efficacy in not only bone regeneration but many other drug delivery applications;

however, the presented work did not demonstrate this ability. Tunable release kinetics are

currently being investigated through modification of various parameters during

electrospinning. These modifications include changes to the solution properties such as the

polymer viscosity and the polymer type or changes to the electrospinning parameters such

as the voltage and flow rate. Modulation of these parameters can be used to alter the fiber

diameter or the degradation rate of the gentamicin sulfate-loaded wrap if tunable release

kinetics are needed following in vivo evaluation [45, 53]. Furthermore, commercialization

of the antimicrobial wrap will require that the fabrication time is reduced. This must be

completed without compromising the effective dose loaded into the wrap. This can be

achieved by using a different solvent such as chloroform to solubilize and load gentamicin

sulfate at higher concentrations [185, 186]. A higher loading concentration will maintain

the requisite total drug load with less polymer being collection over a shorter period of

time. Careful consideration should be given when modifying the loading concentration as

to not increase the gentamicin sulfate concentration above a critical threshold that would

cause drug aggregation [209, 210]. This could result in changes to the release kinetics and

alter the clinical efficacy.

In this work, we have also demonstrated the potential of an engineered substrate to

serve as an adjunct wrap to guide formation of the induced membrane. We have shown that

this wrap provides bimodal release of gentamicin sulfate and VEGF with sustained

bioactivity to both prevent infection and enhance angiogenesis of the induced membrane.

Another proposed key feature of this wrap is the ability to recruit cells and guide cellular

Page 147: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

124

behavior to influence membrane formation with improved durability. However, in vitro

cell studies that evaluate attachment, matrix deposition, proliferation, and migration are

needed to validate this claim. Additional in vivo studies are required to determine the

duration that the wrap retains its structural integrity. Degradation must occur at a rate that

allows for tissue remodeling. If degradation proceeds tissue remodeling, then the fiber

populations can be tuned independently to slow this rate by mechanisms such as altering

the crosslink density of the gel-MA and using a higher molecular weight PLGA or PLGA

with a different co-polymer ratio that is more hydrophobic [290, 312]. In terms of

enhancing angiogenesis, it was observed in in vitro studies that co-electrospinning reduced

the total load of VEGF due to a reduction in fiber collection as compared to the fiber

collection of gel-MA during independent electrospinning. A longer collection time will be

required to overcome the loss of fibers during co-electrospinning and increase the available

load. The overall potential of the multifunctional wrap to guide formation of the induced

membrane will need to be evaluated in vivo to obtain clinically relevant information. It will

be important to observe the tensile properties of the induced membrane as to determine if

the multifunctional wrap improved durability that improves handling. If mechanical

properties are not improved, then the fiber alignment can be modified to guide matrix

deposition and orientation and corollary mechanical properties [54]. However, doing so

may alter mechanotransduction during the second stage of the procedure and impact bone

regeneration. Another important aspect to observe is the vascularity of the induced

membrane. In vivo analysis of the VEGF-loaded gel-MA mesh should be performed

subcutaneously first to confirm the angiogenic potential of the released concentrations. If

Page 148: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

125

vascularization is not enhanced, then the wrap can be optimized by increasing its thickness

to increase the available dose of VEGF. The effect of improved durability and enhanced

vascularization should then be evaluated on bone healing to establish the efficacy of the

multifunctional wrap. This can be achieved with histological analysis, radiographic

analysis, and mechanical analysis. These same observations should made for the wrap in

infected bone defects as to determine the potential to simultaneously prevent infection and

promote membrane formation for improved bone healing in contaminated bone defects.

Despite the need for additional investigation to further develop and confirm the

potential the multifunctional wrap, the work presented here has advanced the wrap towards

the goal of being an adjunct substrate to improve bone regeneration during the Masquelet

technique. First, an electrospun gentamicin sulfate-loaded PLGA wrap was recognized as

an ideal adjunct therapy to treat osteomyelitis. Then the electrospun antimicrobial mesh

was combined with an electrospun VEGF-loaded gel-MA mesh to establish a resorbable

substrate that offers bacterial inhibition, templating for cellular recruitment and matrix

deposition, and biochemical signaling to direct cellular behavior during tissue remodeling.

The findings from these studies provide the fundamentals for further development of the

multifunctional electrospun wrap as well as contribute to the knowledge of electrospun

platforms available for use as stand-alone therapies or combined constructs to meet the

complex requisites for improving tissue regeneration for a variety of applications.

Page 149: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

126

Appendix A: In Vivo Performance of a Bilayer Wrap to Prevent

Abdominal Adhesions 5

A.1 INTRODUCTION

Intestinal anastomoses following bowel resection are among the most frequently

performed procedures in general surgery [313]. Despite advancements in suturing and

stapling techniques, there is a high incidence of post-operative complications with up to

30% of patients presenting with anastomotic leakage and up to 90% of patients developing

intra-abdominal adhesions [314-317]. Anastomotic leakage is of particular concern due to

its associated high morbidity and mortality rates as a result of infection, peritonitis, and

sepsis [316, 318, 319]. Treatment of anastomotic leakage often requires prolonged

hospitalization and a secondary operation to minimize infection. Intra-abdominal

adhesions form vascularized fibrotic bridges between adjacent tissues that can restrict

normal organ movement and blood supply leading to chronic pain and bowel obstruction

[320, 321]. The severity and widespread occurrence of these two complications underscore

the need for a method to improve patient outcomes after these surgical procedures.

The two most prevalent approaches to reduce anastomotic leakage are

reinforcement of the anastomotic closure with surgical sealants (i.e., fibrin and

cyanoacrylate glues) and an adjunct wrap placed over the anastomosis. Despite in vivo

experimental efforts to develop a method using surgical sealants to strengthen the suture

5 This work was reprinted with permission from “In vivo performance of a bilayer wrap to prevent

abdominal adhesions”, by Alysha Kishan, Taneidra Buie, Canaan Whitfield-Cargile, Anupriya Jose, Laura

Bryan, Noah Cohen, and Elizabeth Cosgriff-Hernandez. Acta Biomaterialia, 2020, 115, 116-126. © 2020

Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.

Page 150: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

127

line, results have displayed high variability of outcomes that were attributed to dosage

variations and inconsistencies in application [322-324]. As an alternative, several groups

have evaluated tissue-derived wraps to externally reinforce the anastomosis and enhance

anastomotic healing including patient-specific omentum, bovine pericardium, human

amniotic membrane, and porcine collagen foams [325-330]. Bovine pericardium, in

particular, has been shown to improve wound healing with significant increases in burst

pressures of the treated anastomoses [328]. Although these materials effectively enhance

healing and reduce anastomotic leakage, none have gained commercial success possibly

due to the increased incidence and severity of intra-abdominal adhesions associated with

their use [331, 332].

To address the complications of intra-abdominal adhesions after intestinal

surgeries, several adhesion barriers have been developed and commercialized. Adhesion

barriers act to physically separate the injured tissue from the adjacent viscera during the

critical period of post-operative healing (5-7 days), thereby reducing the formation of

fibrotic adhesions [333]. Seprafilm, composed of sodium hyaluronate and

carboxymethylcellulose, and Interceed®, composed of an oxidized regenerated cellulose,

are adhesion barriers that effectively reduce the incidence and severity of adhesions both

experimentally and clinically [320, 334, 335]. Despite established efficacy, these barriers

have limitations. The efficacy of Interceed® is significantly reduced in the presence of

blood due to fibrin deposition and fibroblast penetration which reduces the capacity to

prevent adhesions [336-338]. Application of Seprafilm® immediately after anastomoses

delays inflammatory-mediated processes that promote healing of the suture line which

Page 151: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

128

leads to an increased rate of anastomotic leakage [337-339]. As a result, several clinical

studies have recommended that surgeons avoid applying Seprafilm directly onto fresh

suture lines [334, 339, 340].

To date, current treatments to enhance anastomotic healing to prevent leakage often

result in increased adhesions; whereas, barrier materials to reduce adhesions can delay

anastomotic healing and increase anastomotic leakage. These findings suggest that

therapeutic strategies that only address one of these complications independent of the other

will ultimately fail. Our laboratory developed a new approach to concurrently address these

complications using a composite bilayer wrap with selective bioactivity to both reduce

intra-abdominal adhesions and enhance anastomotic healing. The inner layer consists of an

electrospun gelatin mesh to promote cell adhesion and remodeling [341]. Reactive

electrospinning developed in our laboratory was utilized to modulate the degree of gelatin

crosslinking and the corollary resorption rate. In order to resist adhesion formation, a

resorbable poly(ethylene glycol) (PEG) hydrogel was selected as an outer layer due to its

inherent non-fouling properties [342]. Iterative design of hydrogel and electrospun mesh

formulations was utilized to identify a bilayer structure with requisite surgical handling

characteristics and retention of wrap integrity after swelling. Candidate bilayer wraps were

then evaluated to confirm selective bioactivity over two weeks. As an in vivo proof-of-

concept study, adhesion prevention was assessed using a rat colonic abrasion model in

comparison to a clinical control using Interceed. Based on initial in vivo results, the

bilayer wrap was further modified to include an additional tissue adhesive component to

prevent displacement after implantation. Collectively, these studies provide an initial

Page 152: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

129

evaluation of a new bilayer wrap design to address the multimodal complications

associated with intestinal anastomoses.

A2. MATERIALS AND METHODS

Materials

All chemicals were purchased from Sigma Aldrich (Milwaukee, WI) and used as

received unless otherwise noted.

Polymer Synthesis

Poly(ethylene glycol) diacrylate (PEGDA) was synthesized according to a method

adapted from Hahn, et al. [343]. Briefly, acryloyl chloride was added dropwise to a solution

of PEG 2 kDa or 6 kDa diol and triethylamine in dichloromethane (DCM) under nitrogen.

The molar ratio of PEG, acryloyl chloride, and triethylamine was 1:2:4, respectively. After

the addition of acryloyl chloride, the reaction was stirred for an additional 24 hours at room

temperature. The resulting solution was then washed with 8 molar equivalents of 2 M

potassium bicarbonate to remove acidic byproducts. The product was then precipitated in

cold diethyl ether, filtered, and dried under vacuum.

PEGDA with thio-β esters (PEGDTT) was synthesized as previously described

[342]. d,l-dithiolthreitol (DTT) and triethylamine (TEA) were added dropwise to a solution

of PEGDA (2 kDa) in DCM. The molar ratio of DTT, PEG and triethylamine was 3:2:0.9,

respectively. After the addition of the DTT and triethylamine, the reaction was stirred for

24 hours at room temperature. The resulting solution was then precipitated in cold

Page 153: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

130

diethethyl ether, washed, filtered, dried under ambient conditions for 24 hours then placed

under vacuum to remove any excess solvent.

Characterization of PEGDA and PEGDTT was confirmed using proton nuclear

magnetic resonance (1H-NMR) spectroscopy. Proton NMR spectra of control and

functionalized polymers were recorded on a Mercury 300 MHz spectrometer using a

TMS/solvent signal as an internal reference. Percent conversions of PEG diol to acrylate

endgroups was greater than 85%. 1H-NMR (CDCl3): δ 3.6 ppm (m, -OCH2CH2), 5.8 ppm

(dd, -CH=CH2), 6.1 (dd, -CH=CH2) and 6.4 ppm (dd, -CH=CH2).

Reactive Electrospinning of Gelatin

In situ crosslinking of electrospun gelatin was performed as previously reported

[341]. Briefly, a double-barrel syringe with an attached mixing head and a diisocyanate

crosslinker were utilized to generate electrospun scaffolds that crosslink during the

electrospinning process via reaction of the isocyanate with the pendant amines of lysine

residues in gelatin. Bovine-derived gelatin, 1,4-diazabicyclo[2,2,2]octane (DABCO) and

hexamethylene diisocyanate (HDI) were each dissolved in 2,2-trifluoroethanol (TFE). The

concentration of HDI was determined such that the crosslinker density would equal a 5:1

ratio of isocyanate:amine. Double-barrel syringes were loaded with 12 wt% gelatin in TFE

and 5 wt% (of solids) DABCO solution in one barrel and HDI in TFE solution in the other.

The double barrel syringe solutions were pumped through a mixing head into an 18-gauge

blunted needle at a rate of 3.0 mL/hr. The needles were placed 12 cm away from the

collector to allow for adequate solvent evaporation and fiber drying. A high voltage of 10

Page 154: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

131

kV was then applied to the needle. Meshes were electrospun for 3 hours and stored under

vacuum for at least 24 hours prior to further use.

Bilayer Wrap Fabrication

A PEG-based hydrogel coating method was selected through iterative design using

3 distinct methods. Namely, a bulk hydrogel coating, an electrosprayed hydrogel coating,

and a hydrogel foam coating were tested. Bulk hydrogel coatings were fabricated from a

10 wt% PEGDTT:PEGDA 75:25 solution in distilled water. A photo-initiator solution (1

mg Irgacure 2959 per 10 µl 70% ethanol) was added at 1 vol% of precursor solution. The

PEG hydrogel precursor solution was pipetted between 1-mm spacer plates and exposed to

UV light to initiate crosslinking. After 45 seconds of UV illumination, a 0.2-mm thick

gelatin mesh was placed on top of the surface of the hydrogel. Samples were then exposed

to UV light for 5 additional minutes to complete hydrogel formation. Bilayer wraps were

swollen in reverse osmosis water overnight followed by lyophilization before further use.

Electrospraying precursor solutions were prepared from 30 wt% 75:25

PEGDTT:PEGDA in 70:30 DCM:EtOH solvent. A photo-initiator solution (1 mg Irgacure

2959 per 0.01 ml 70% ethanol) was added at 3 vol% of precursor solution. The hydrogel

solution was then added to a syringe and electrospun gelatin meshes were attached to a

copper plate as the collector. A voltage of 7.5 kV was applied to the needle tip and -2 kV

was applied to the collector, which was set 13 cm away from the needle tip to allow for

solvent evaporation and particle drying. The polymer solution was dispensed at a rate of

1.0 mL/hr using a syringe pump. A UV lamp was placed above the electrospraying set up

Page 155: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

132

to enable in situ curing. The hydrogel solution was electrosprayed for 3 hours, after which

the resulting bilayer wrap was vacuumed for at least 24 hours prior to characterization.

Finally, PEG-based hydrogel foam coatings were prepared using an emulsion

templating method developed previously in our laboratory. Hydrogel precursor solutions

were prepared by mixing 20 wt% PEGDTT:PEGDA 75:25 in a 10 wt% solution of

Pluronics F-68 in water, and a photo-initiator solution added at 1% of the total polymer of

lithium phenyl-2,4,6-trimethylbenzoylphosphinates (LAP). Precursor solutions were made

with and without 10 wt% trimethyloylpropane ethyodylate triacrylate (TMPE-TA) as an

additional crosslinker. Once thoroughly mixed, light mineral oil was added at volume

fractions of 50% of the continuous polymer precursor phase. The water and oil composition

was mixed at 2500 rpm for 5 minutes using a FlackTek Speedmixer DAC 150 FVZ-K.

Electrospun gelatin meshes were affixed to 1.0- mm spacer plates and hydrogel emulsions

were then transferred into the mold. Samples were exposed to UV light for 6 minutes to

complete foam formation. Wraps were then subjected to a series of 3 1-hour soaks to

remove the oil content: 1) 100% DCM; 2) 50/50 DCM/ethanol; 3) 100% ethanol. After the

last ethanol soak, wraps were soaked in distilled water overnight prior to lyophilization

before further use.

Bilayer Wrap Characterization

Scanning electron microscopy (SEM) (JOEL 6500) was utilized to image

specimens. Lyophilized samples were stored under vacuum prior to imaging. A total of 4

composite bilayer wraps were fabricated for each composite formulation. One specimen

Page 156: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

133

was taken from each wrap to yield 4 specimens/group. Specimens were coated with 4 nm

of gold using a sputter coater (Sputter Coater 108, Cressingtion Scientific Instruments).

Confirmation of two distinct layers was performed by evaluating the chemical structure of

each using attenuated total reflectance-Fourier transform infrared (ATR-FTIR)

spectroscopy (Nicolet iS10 (Thermo Scientific) at a resolution of 2 cm-1 for 32 scans.

A previously described, 4 composite bilayer wraps were fabricated for each

composite formulation. One dog-bone specimen was cut from each wrap to yield 4

specimens/group in accordance with ASTM D1708. Specimens were strained to failure at

a rate of 100 %/min based on the initial gauge length using an Instron 3345 uniaxial tensile

tester equipped with a 100-N load cell and pneumatic side action grips (Instron 2712-019).

The elastic modulus, tensile strength, and ultimate elongation were calculated from the

resultant engineering stress-strain curves. A secant modulus at 2% strain was calculated

for the elastic modulus and subsequently referred to as “modulus”. Toughness was

calculated as the area under the stress-strain curve.

Selective bioactivity over time was assessed via cell adhesion. Adult human dermal

fibroblasts (hDFs, isolated from adult skin, Invitrogen) were cultured in growth media

containing 16.5% fetal bovine serum (FBS, Atlanta Biologicals), 1% L-glutamine (Life

Technologies), and Minimum Essential Media α (MEM α, Life Technologies) to 80%

confluence and utilized at Passage 5. A total of 4 hydrogel foam fiber composite bilayer

wraps were fabricated and 8 mm-disks were removed from each wrap using a biopsy punch

(Integra Miltex). Specimens were placed in a tissue cultured 48-well plate (Corning) and

were sterilized for 30 minutes under UV. They were then immersed in PBS and incubated

Page 157: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

134

at 37 °C for 2 weeks. PBS was selected as the hydrogel component is hydrolytically

degradable and incubation in PBS is a common analogue for real-time hydrolytic

degradation under physiological conditions (e.g. pH and ion concentration) [344-346].

Specimens (n=4/group) were then taken at 1, 7, and 14 days to test selective bioactivity.

Human dermal fibroblasts were seeded upon either the hydrogel foam layer or the gelatin

layer at 10,000 cells/cm2 in growth media supplemented with 1 vol% penicillin-

streptomycin. Following a 3-hour incubation, cells were fixed and stained with rhodamine

phalloidin (F-actin/cytoplasm, Biotium) and SYBR green (DNA/nucleus, Thermo Fisher

Scientific). A 3-hour culture period was selected as it is considered a typical length of time

to allow cell attachment [347-349]. Cell adhesion was calculated from manual cell counts

of images obtained through raster patterning of 4 specimens/group/end point using a

fluorescence microscope (Nikon Eclipse TE2000-S).

Animal Care

All procedures were approved by the Texas A&M University Institutional Animal

Care and Use Committee (IACUC, Animal Use Protocol# 2017-0316).

In Vivo Assessment

First, in vivo resorption rates for each component of the bilayer mesh was assessed.

Sprague-Dawley rats (n = 3/group) were randomly assigned to 1 of 2 groups: 1) hydrogel

foam or 2) electrospun gelatin mesh to assess degradation. Animals were anesthetized with

isoflurane in oxygen and maintained on isoflurane in oxygen via a nose cone. The ventral

Page 158: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

135

abdomen was clipped and prepped for surgery with chlorohexidine and 70% ethanol. The

abdomen was draped with a sterile paper drape and a 2-cm ventral mid-line incision made

in the central third of the abdomen. The appropriate dried mesh or gel was paced in the

abdomen in a sterile manner. The body wall was closed with 5-0 PDS in an intradermal

pattern and the skin was closed with 5-0 PDS in a simple continuous pattern. A

circumferential abdominal bandage was placed and all animals were administered

buprenorphine (0.01 mg/kg, subcutaneously) immediately post-operatively. At each time

point, the rats were euthanized via carbon dioxide asphyxiation. The abdomen was opened

and entire abdomen thoroughly explored macroscopically to identify remaining specimens

or confirm full resorption.

The effect of treatment on the formation of intra-abdominal adhesions was assessed

using a rat cecum abrasion model as previously described [350]. Sprague-Dawley rats (n =

5/group) were randomly assigned to 1 of 6 groups: 1) cecum abrasion with no treatment;

2) cecum abrasion with fibrin glue applied; 3) cecum abrasion with Interceed ® applied;

4) cecum abrasion with gelatin mesh only applied; 5) cecum abrasion with hydrogel foam

only applied; 6) cecum abrasion with bilayer wrap applied. Specimens were sterilized using

ethylene oxide and allowed to vent for 3 days prior to use. Twelve-week-old rat were

anesthetized and prepared for surgery as described above. A 2-cm ventral mid-line incision

made in the central third of the abdomen. The cecum was exteriorized and a template used

to abrade the cecal serosa in a standard location, size (1 x 2 cm2), and severity of abrasion

(40 strokes of a dry gauze with mild pressure). Similarly, a 1 x 2 cm2 area of body wall

was resected from the right side of the abdomen centered 1 cm dorsal to the incision. A

Page 159: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

136

single simple interrupted suture (5-0 monocryl) was placed between the cecum and the

body wall at a site approximately 1 cm distant (cranial) to the abraded area of both the

cecum and body wall. The treatments were then applied. Briefly, Tisseel® tissue sealant

was used as fibrin glue (Baxter healthcare) and was thawed in a water bath 1 hour prior to

use and used within 4 hours of thawing. Approximately 200 µL of fibrin glue was applied

to the abraded surface of the cecum for every group except PBS and Interceed®. The

appropriate mesh was then immediately applied to the area making every effort to cover

the abraded area with the mesh. For Interceed®-treated rats, fibrin glue was not applied

and instead Interceed® was applied according to manufacturer’s directions (applied dry to

a non-bleeding surface). Similarly, approximately 500 µL PBS was pipetted onto the

damaged portion of the cecum 3 times for the PBS group. The cecum was then returned to

the abdomen. The body wall was closed with 5-0 PDS in a simple continuous pattern and

the skin was closed with 5-0 PDS in a simple continuous pattern. A circumferential

abdominal bandage was placed and all animals were administered buprenorphine (0.01

mg/kg subcutaneously) immediately post-operatively.

At each time point, animals were euthanized via carbon dioxide asphyxiation. The

abdominal wall was opened at the left flank to avoid disturbing site of adhesion and

retracted to expose the right body wall. Adhesions were photographed and scored discretely

for size, strength, and maturity as described in Table A.2. One observer blinded to

treatment group scored all of the animals. As the small scale used to determine adhesion

scores (0-3, 0-5) does not capture the differences between large, mature adhesions, and

Page 160: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

137

smaller, less cohesive adhesions, a composite score was assigned to each rat based on the

following calculation:

𝐶𝑜𝑚𝑝𝑜𝑠𝑖𝑡𝑒 𝑠𝑐𝑜𝑟𝑒 = 𝑠𝑖𝑧𝑒 × (𝑠𝑡𝑟𝑒𝑛𝑔𝑡h + 𝑚𝑎𝑡𝑢𝑟𝑖𝑡𝑦)

Dopamine Tissue Adhesive Coating

Dopamine hydrochloride was dissolved at 50 mg/mL in 10 mM tris. 1 mL of

solution was then carefully pipetted onto the gelatin layer of a swollen bilayer wrap. After

24 hours, the solution was aspirated and 1 mL of 3 wt% sodium periodate in distilled water

was applied to the bilayer wrap. After 1 minute, the sodium periodate solution was

aspirated and wraps were washed in 50 mL of distilled water for 5 hours with water changes

every hour. Samples were subsequently frozen and lyophilized prior to characterization.

The dopamine-modified gelatin layer was affirmed using ATR-FTIR spectroscopy at a

resolution of 2 cm-1 for 32 scans.

Adhesion Strength Measurement

Adhesive properties of the dopamine-coated gelatin layer were determine using lap-

shear tensile stress measurements in accordance with the ASTM standard F2255-05. Fresh

split thickness porcine skin was cut into 2.5 x 5 cm2 segments with uniform thickness and

hydrated in PBS for immediate use. A total of 4 un-modified and adhesive gelatin meshes

were fabricated and stored in a dry location at room temperature. A 2.5 × 1 cm2 specimen

was removed from each mesh to account for batch variability. Interceed® (Ethicon) sheets

were cut into comparable dimensions for comparison to the un-modified and adhesive

Page 161: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

138

gelatin meshes. Interceed® was selected as a clinical control due to its ability to securely

adhere to the anastomosis without sutures when meticulous hemostasis has been achieved

[336]. The porcine test substrates were removed from PBS and blotted with sterile gauze

to remove excess PBS prior to coming into contact with the un-modified gelatin specimens,

adhesive gelatin specimens, or Interceed®. The specimens were positioned between two

tissue substrates with a 1 cm overlap. They were then compressed with a force of

approximately 1 N to allow for the bond to set. Specimens were further conditioned in a

humidity chamber at 37°C for 30 minutes prior to being strained to failure at a rate of 5

mm/min using an Instron 3345 uniaxial tensile tester equipped with a 100-N load cell and

pneumatic side action grips (Instron 2712-019). The maximum strength and failure strain

were recorded. The adhesive strength was calculated by maximum strength divided by the

initial bond area.

Statistical Analysis

Data are displayed as mean ± standard deviation for each composition. Statistical

analysis was performed utilizing a standard one-way ANOVA unless otherwise noted in

the figure captions. Statistical significance was accepted at P< 0.05.

A.3 RESULTS

Bilayer Wrap Optimization

Page 162: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

139

Multiple bilayer formulations were investigated for their suitability to produce an

optimized adhesion barrier with enhanced handleability and stability in vitro. The proposed

PEG-based hydrogel layer for adhesion prevention was applied to the inner electrospun

gelatin layer in 3 forms: a hydrogel layer, an electrosprayed layer, and a foam layer (Figure

A.1A-C). Each of the 3 bilayer wrap fabrication methods were assessed for handling in

terms of tensile properties and stability via delamination assessment upon swelling. The

hydrogel bilayer wrap displayed the most brittle behavior, as evidenced by the stress-strain

curves in Figure A.1C. The electrosprayed bilayer wrap had the lowest toughness (0.4 ±

0.2 MJ/m3) and reached tensile failure at 3.5 ± 1.2% elongation (Table A.1). The hydrogel

foam composite bilayer wrap displayed the highest toughness of 23.6 ± 1.7 MJ/m3 and

reached an elongation of 41.0 ± 4.0%. As such, this composite formulation was selected

for future testing. However, swelling analysis indicated delamination upon immersion in

water. In order to decrease the degree of swelling and thereby resist delamination, the

crosslinker, trimethylolpropane ethoxylate triacrylate (TMPE-TA), was added to the foam

precursor solution. The TMPE-TA containing bilayer was again evaluated to ensure

maintenance of handleability and stability in vitro. The formulation displayed a further

significant increase in toughness, from 23.6 ± 1.7 to 28.8 ± 1.5 MJ/m3 (Table A.1).

Furthermore, the formulation displayed no evidence of delamination upon swelling. This

optimized bilayer wrap, containing an electrospun gelatin layer and PEG-TMPETA foam

was subjected to further characterization prior to assessment in vivo.

Page 163: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

140

Figure A.1: Iterative design of a composite bilayer wrap with requisite mechanical

properties. A) Schematic of composite bilayer wrap fabrication for bulk hydrogel-fiber

composite, electrosprayed hydrogel-fiber composite, and hydrogel foam-fiber composite.

B) Set up for mechanical testing of the three composite formulations and C) resulting

stress-strain response for each composite bilayer wrap (arrows indicate tensile failure at

the associated strain). *Figure created with BioRender.com.

Page 164: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

141

Table A.1: Tensile properties of composite bilayer wrap formulations. * indicates

statistical differences as compared to the hydrogel foam-fiber composite, (P<0.05).

Composite

formulations

2% Secant

modulus

(MPa)

Ultimate tensile

strength (MPa)

Elongation

(%)

Toughness

(MJ/m3)

Bulk hydrogel -

fiber composite 22.8 ± 4.8 * 0.6 ± 0.2* 2.5 ± 0.5 * 1.0 ± 0.4 *

Electrosprayed

hydrogel - fiber

composite

4.8 ± 0.7 0.1 ± 0.1 * 3.5 ± 1.2 * 0.4 ± 0.2 *

Hydrogel foam

-

fiber composite

4.7 ± 0.7 0.7 ± 0.1* 41.0 ± 4.0 * 23.6 ± 1.7 *

Hydrogel foam

+ TMPETA -

fiber composite

4.1 ± 0.2 0.9 ± 0.1* 48.3 ± 3.4 * 28.8 ± 1.5 *

Bilayer Wrap Characterization

ATR-FTIR spectroscopy was utilized to evaluate the chemical composition of each

layer in the selected bilayer wrap. Spectral analysis indicated that although the hydrogel

was visible on both layers, as demonstrated by the ether peak at 1110 cm-1, the amine

stretch at 3350 cm-1 of gelatin was only present on the inner layer (Figure A.2A). Scanning

electron microscopy of the selected bilayer wrap, displayed in Figure A.2B, demonstrated

that the inner gelatin layer consisted of homogenous, smooth fibers while the outer

hydrogel foam layer displayed a porous intra-architecture encased between the fibrous

layer and a smooth, outer film.

Page 165: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

142

Figure A.2: Characterization of the hydrogel foam + TMPETA- fiber composite. A)

ATR-FTIR of the gelatin layer (gray line) and the hydrogel layer (black line). B) Cross-

sectional SEM depicting the intra-microarchitecture of each layer (scale bar =30 µm).

SEM blowouts display the plan view of each layer (scale bar =100 µm). C)

Representative images of hDF attachment over 14 days (scale bar =200 µm) and

quantified cell adhesion on each layer. Cells stained with rhodamine phalloidin (F-

actin/cytoplasm) and SYBR green (DNA/nucleus). * indicates statistical differences with

respect to the hydrogel foam at each time point, (two-way ANOVA with Sidak’s

analysis, P<0.05).

Next, the potential to maintain selective bioactivity over the desired application

period (target 2 weeks) was evaluated by assessing hDF adhesion on each layer at selected

Page 166: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

143

time intervals. Bilayer wrap samples were incubated in PBS for 2 weeks and removed at

1, 7, and 14 days to prior to culturing with cells to determine the effect of degradation on

bioactivity. After each time point, hDFs were seeded onto each side of the bilayer wrap for

3 hours and cell adhesion was evaluated. There was significant cell adhesion on the

electrospun gelatin layer throughout the 14-day period, (Figure A.2C). In contrast, the anti-

fouling properties of the hydrogel foam layer prevented cellular adhesion resulting in cells

being washed away prior to analysis. Overall, these results suggested that selective

bioactivity was maintained.

Assessment of In Vivo Degradation

Prior to evaluating the efficacy of the bilayer wrap, an in vivo degradation scouting

study was conducted. Samples of each composition were implanted within the abdominal

cavity of rats and degradation was assessed visually after 7, 14, and 21 days. Initial studies

indicated that gelatin meshes were present at 7 days and had lost mechanical integrity by

14 days. Hydrogel foam specimens remained at 21 days, suggesting that a longer study

must be conducted to fully evaluate the in vivo degradation rate. These initial results

indicated that the degradation profiles of each component were within range of the targeted

rate, permitting initial evaluation for adhesion prevention.

In Vivo Evaluation of Intra-abdominal Adhesion Formation

Intestinal adhesion formation between the cecum and peritoneum of rats was

evaluated by observation of abdominal wound sites within a rat abrasion model after 14

Page 167: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

144

days (Figure A.3). Initial scouting studies indicated that applying the bilayer wrap to the

wound site with sutures resulted in strong adhesion formation initiated by leakage, and

ultimately infection. As a result, fibrin glue was used to affix the bilayer wrap to the

abraded area. The adhesions were evaluated macroscopically to discretely evaluate their

area, strength, and maturity (data not shown). A summary of the adhesion scoring method

is described in Table A.2. In addition to the adhesion scoring system, a composite score

was also calculated in an attempt to better capture the differences in adhesions by

considering the correlation between size, strength, and maturity. Figure A.3B displays a

summary of the composite scores as well as representative images from each group (Figure

A.3C-H). Control rats, and rats with only fibrin glue application, showed the development

of strong, thick adhesions between the cecum and peritoneum in all instances, with

composite scores of 18 ± 9 and 20 ± 4, respectively. Interceed® was utilized as a clinical

control and showed complete prevention of adhesions in all specimens (composite score =

0 ± 0). Gelatin meshes applied alone saw a range of adhesion formation in the rats, with a

composite score of 12 ± 10 that was statistically greater than Interceed®. This was expected

as gelatin is known to promote cell adhesion, which the bilayer aims to utilize to promote

healing. Hydrogel foam control and bilayer wrap treated rats had composite scores of 8 ±

7 and 8 ± 5 each. Though these scores were higher than that for Interceed® treated rats,

they were not statistically different. The higher scores were attributed to a subset of rats in

both groups presenting with immature adhesions at the wound site. It was noted that the

foam and bilayer wraps had been displaced from the wound site in each of the rats where

adhesions were reported. Few adhesions were found in rats where the foams or wraps

Page 168: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

145

remained in place. As such, it was concluded that the fibrin application was insufficient to

maintain placement and that wraps needed to be formulated such that they adhered to the

wound bed for further studies.

Page 169: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

146

Figure A.3: In vivo intra-abdominal adhesion study results. A) Schematic of rat in vivo

abrasion model utilized to assess adhesion formation. B) Composite adhesion scoring for

all treatments, red data points indicate observed displacement of the treatment specimen.

* indicates statistical differences as compared to Interceed®, (P<0.05). Representative

images taken from site where treatments were applied: C) PBS treated, D) Tisseel®

treated, E) gelatin mesh treated, F) hydrogel foam treated, G) composite bilayer treated,

H) Interceed® treated. *Figure created with BioRender.com

Table A.2: Adhesion scoring description.

Area Score

0 No Adhesion

1 Cecum to bowel adhesion

2 Cecum to body wall (<25% of abraded area)

3 Cecum to body wall (25-50% of abraded area)

4 Cecum to body wall (50-100% of abraded area)

5 Cecum to body wall (>100% of abraded area)

Strength Score

0 No adhesion

1 Gentle traction required to break adhesion

2 Blunt dissection required to break adhesion

3 Sharp dissection required to break adhesion

Gross Adhesion Maturity Score

0 No adhesion

1 Filmy adhesion

2 Vascularized adhesion

3 Opaque or cohesive adhesion

Development of a Tissue-adhesive Bilayer Wrap

Dopamine modification of the electrospun gelatin was investigated as a method to

confer adhesive properties to the bilayer wrap to prevent displacement. Electrospun gelatin

Page 170: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

147

meshes were coated with dopamine through the reported dip-coating method (Figure

A.4A). Dopamine-modified gelatin meshes were then oxidized and lyophilized to preserve

their oxidized state. Infrared spectral analysis indicated the presence of the dopamine-

specific peaks in the modified gelatin meshes (Figure A.5). Lap shear testing was utilized

to evaluate the adhesive properties of the dopamine-modified gelatin mesh as compared to

un-modified gelatin, Interceed®, and fibrin glue. Specimens were applied to split-thickness

porcine skin and subjected to tensile testing as depicted in Figure A.4B. The mean shear

strength of the dopamine-modified electrospun gelatin layer was 11.6 ± 2.9 kPa. This value

is significantly higher than values reported for fibrin glue (4.6-6.9 kPa) and comparable to

that obtained from commercial Interceed® (Figure A.4C) indicating that dopamine can

potentially prevent displacement [304, 351]. The shear strength of the un-modified gelatin

mesh was not included as, compared to other groups, the hydrated gelatin wrap had shear

strength associated with forces that were below the detection limit of the load cell.

Page 171: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

148

Figure A.4: Effect of dopamine coating on tissue adhesion to the bilayer wrap. A)

Schematic of dopamine coating process and hypothesized reaction of dopamine coating

with the wound site. B) Schematic depicting the lap shear test set-up and C) average

maximum shear strength of the dopamine coated bilayer wrap on porcine substrates as

compared to Interceed®. Un-modified gelatin control was not included in graph due to

shear forces below the detection limit of the instrument. * indicates statistical differences

as compared to Interceed®, (student’s t-test, P<0.05). *Figure created with

BioRender.com.

Page 172: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

149

Figure A.5: Characterization of the dopamine-modified gelatin mesh. A) ATR-FTIR of

the un-modified gelatin (black line), pure dopamine (dark gray line), and dopamine-

modified gelatin mesh (light gray line).

A.4 DISCUSSION

It remains a challenge to improve healing of intestinal anastomoses complicated by

anastomotic leakage. Current treatments to improve anastomotic healing often result in

increased adhesions [19, 26]. Adhesion development typically occurs within the first 5 days

after surgery and is driven by the natural healing cascade. After damage at the peritoneum,

macrophages and mesothelial cells secrete cytokines and other inflammatory mediators to

initiate re-epithelialization [352]. During this process, fibrin is deposited at the injured site,

forming a matrix to facilitate repair. At this time, fibrin bridges may form between two

adjacent damaged surfaces. Typically, fibrin is then broken down by fibrinolytic enzymes

leaving behind a healthy mesothelial layer. However, the activity of these enzymes can

Page 173: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

150

become compromised due to surgical damage limiting fibrin breakdown. This results in an

influx of fibroblasts, capillaries, and nerves that lead to the development of permanent

fibrous connective tissue, also known as adhesions [353]. Adhesion barriers such as

Seprafilm® and Interceed® typically prevent adhesion formation by physical separation

between tissues during the first 5 days. Contrarily, there is concern with the use of adhesion

barriers as adhesion formation has an integral role in the 1 to 2 week critical re-

epithelialization phase of the anastomotic healing [338, 354]. Prevention of adhesion

formation has been shown to delay anastomotic healing leading to increased incidences of

leakage [331, 338, 354]. Thus, barrier films that solely address adhesion prevention fail to

holistically address the clinical needs and can lead to more severe complications associated

with anastomotic leakage. Key requirements for an improved adhesion barrier include that

it promotes anastomotic healing, is simple to apply, is stable in aqueous environments,

degrades within the targeted time-frame, and maintains its position during treatment [355].

To this end, we have developed a resorbable bilayer wrap that resists adhesion

formation while maintaining the potential to promote healing of the anastomosis site. The

inner layer consists of a crosslinked gelatin electrospun mesh. Gelatin, a natural polymer,

inherently promotes cellular adhesion through its RGD (arginine-glycine-aspartate) ligand

[298]. The fibrous structure of the electrospun mesh also promotes cellular integration by

mimicking the native structure of the extracellular matrix [356]. As the gelatin layer of the

bilayer wrap is present to promote this re-epithelialization, the targeted degradation time

for the wrap is 2 weeks to provide a buffer for delayed healing. Previous work demonstrates

a methodology to crosslink gelatin during the electrospinning process with a diisocyanate

Page 174: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

151

for improved fiber morphology retention and controlled enzymatic degradation. Enzymatic

degradation facilitates cleavage of peptide bonds within the gelatin structures resulting in

resorption byproducts that are cytocompatible and cleared from the body [341, 357]. In

vitro studies demonstrated that the gelatin layer reached full degradation in enzymatic

solution by 14 days [341]. Furthermore, the outer layer of the wrap is a degradable PEG-

based hydrogel foam. PEG-based hydrogels have an intrinsic resistance to cell adhesion

and thereby have the potential to prevent adhesion to surrounding tissue. This was

demonstrated by West et al. who utilized an in situ crosslinked polyethylene glycol-co-

lactic acid diacrylate hydrogel to effectively reduce the adhesion formation by 60% in a

similar rat model [358]. Although adhesion formation can be prevented during the first 5

days by the PEG-based foam, it was hypothesized that the PEG layer degradation rate

should be slower than that of the gelatin layer (targeted 3 weeks). PEGDA degradation is

primarily mediated by hydrolysis of the esters within the acrylate endgroups. As there are

low numbers of esters present in PEGDA, the in vivo degradation rate is slower than desired

[344, 359]. A modified PEGDA-based foam with thio-β ester linkages was utilized to

control hydrolytic degradation to achieve a targeted degradation rate. The incorporation of

thio-β ester linkages increases the positive atomic charge on the carbonyl carbon of

proximal acrylate esters bonds which heightens ester hydrolysis via nucleophilic reactivity.

Higher stoichiometric monomer ratios of thio-β ester linkages to PEGDA results in more

hydrolytically labile sites leading to faster degradation [360]. Previous in vitro work

demonstrated that the modified PEG hydrogel degraded via hydrolysis within 3 weeks and

that degradation byproducts were cytocompatible [342, 360].

Page 175: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

152

In contrast to other PEG-based formulations, we hypothesized that the PEG foam

would provide the requisite toughness and elongation for improved handleability and ease

of application of the bilayer wrap. The favorable mechanical properties of the foam-based

bilayer wrap were attributed to the foam’s microarchitecture (i.e., porosity and pore size)

which were consistent with mechanical properties of hydrogel foams possessing the similar

microarchitecture [361, 362]. It has been noted that hydrogel-based foams with similar

porosity and pore sizes have relatively lower tensile strength and modulus with corollary

enhanced elongation [362]. Equally important to the ease of application is the stability of

the bilayer wrap in terms of delamination. The bilayer wrap displayed delamination which

was attributed to appreciable hydrogel layer swelling. TMPE-TA was added to the

hydrogel foam precursor solution as it is well established in literature that the addition of a

crosslinker reduces scaffold swelling [363]. The addition of TMPE-TA further increased

the strength of the bilayer wrap improving handleability and making it more conducive for

clinical application.

A common drawback of commercial adhesion barriers is that they compromise

anastomotic healing. An advantage of the bilayer wrap is that is has distinct layers to impart

selective bioactivity. Although ATR-FTIR analysis indicated that hydrogel was present on

both layers, the presence of hydrogel on the inner layer does not preclude the cell adhesive

characteristics of gelatin [364]; therefore, these results were considered to be acceptable.

Fibroblasts were selected for evaluation of selective bioactivity as they are the most

prevalent cell type during the healing process and are commonly utilized as a screening

method for adhesion evaluation [365]. Cell adhesion primarily on the gelatin layer

Page 176: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

153

indicated that cell adhesion was unaffected by the presence of a minuscule amount of PEG

foam. This was expected as gelatin is widely incorporated into PEG-based scaffolds to

enhance cellular adhesion [366, 367]. The presence of hydrogel on the gelatin layer is

preferred over the presence of gelatin on the hydrogel layer as the gelatin would reduce the

adhesion resistant properties of the hydrogel layer [364].

In vivo evaluation of the bilayer wrap in a rat abrasion model demonstrated that the

selected compositions of gelatin and PEG-based hydrogel foam were within the targeted

degradation time-frame. The bilayer wrap resulted in reduced adhesion formation. Further

reduction, closer to that of Interceed®, is expected provided that there is maintenance of

the position of the bilayer wrap relative to the wound site. Wraps noted as being displaced

resulted in minor adhesions and indicated a need to more securely affix the wraps to the

wound. Recent studies have reported that dopamine layers can be added to substrates in

order to impart tissue adhesive properties through simple dip-coating [304]. This

technology is derived from mussels, which are known to adhere to all types of organic and

inorganic surfaces under wet conditions. Dopamine hydrochloride (dopamine) has been

identified as the compound which facilitates mussel adhesion to substrates through both

covalent and noncovalent interactions [368]. As a result, dopamine has become a popular

candidate to promote tissue adhesion in devices [369]. We hypothesized that a dopamine

coating could facilitate crosslinking with the wound surface as the catechol transitions into

a quinone upon oxidation, which can then form a covalent bond with primary amines

through nucleophilic attack [351, 370]. This reaction would permit adhesion between the

dopamine-coated gelatin layer and the wound surface. In vitro evaluation of the adhesive

Page 177: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

154

gelatin layer demonstrated marked tissue adhesion as compared to that of un-modified

gelatin, comparable to that of Interceed®, and higher than adhesion strengths reported for

fibrin glue [337-339]. These findings indicate that the adhesive dopamine coating can be

implemented to effectively prevent displacement of the bilayer wrap during healing to

prevent adhesion formation.

Overall, these unique wraps utilize material properties to maintain selective

bioactivity throughout degradation in a controlled manner. By combining the native

properties of PEG hydrogels and gelatin, this bilayer approach could be used to

simultaneously enhance anastomotic healing while preventing abdominal adhesions,

unlike any current clinical standard. As a first step towards this goal, the presented studies

provide an initial evaluation of the bilayer wrap with optimized handling, controlled

degradation, and tissue adhesiveness. Despite promising initial results, there were

limitations in the current study that necessitate future research. Regarding the in vitro

characterization, only hydrolytic degradation was considered during evaluation of the

selective bioactivity despite enzymatic degradation being the primary mechanism of

gelatin degradation. In vivo evaluation of gelatin control meshes did confirm that

enzymatic degradation would not impede the cell instructive behavior of gelatin as

adhesions readily formed over 2 weeks. In addition, future studies are needed that provide

in vitro characterization of the dopamine-modified bilayer wrap to ensure that the

mechanical properties and selective bioactivity is maintained. The adhesive strength of

dopamine-modified gelatin meshes was also not experimentally evaluated against fibrin

glue but the shear strength of fibrin glue is well documented in the literature using the

Page 178: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

155

ASTM F2255 – 05 standard. Following validation of the dopamine-modified bilayer wrap,

prevention of intra-abdominal adhesion with the dopamine adhesive-modified bilayer wrap

will need to be evaluated in vivo. Most notably, the current studies did not evaluate the

efficacy of the bilayer wrap to promote anastomotic healing. The demonstration of reduced

adhesions from this study using an abrasion model provides the requisite proof of principle

for future study in the more rigorous anastomoses animal model. These future animal

studies will need to assess anastomotic healing by evaluating the tensile strength and burst

strength of the treated anastomoses as well as collagen deposition and organization at the

site. Moreover, results will need to be compared to Interceed® in order to assess

improvement upon clinical standards. The full in vivo degradation rate and

biocompatibility assessment of each layer of the bilayer wrap will also need to be

evaluated. If the degradation profile significantly differs from our initial scouting, the rates

may be adjusted according to crosslink density (gelatin layer) and DTT content (hydrogel

layer). Collectively, this tunable system has the potential to have a direct impact upon

patients through rational design of a more effective adhesion barrier with the capacity to

enhance anastomotic healing.

A.5 CONCLUSIONS

In this study, we successfully fabricated a bilayer wrap that retains the advantages

of each respective material to form a composite device for the simultaneous treatment of

adhesion formation and anastomotic healing. Specifically, the completed studies establish

initial efficacy to prevent surgical adhesions with the potential to enhance anastomotic

Page 179: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

156

healing. This wrap has been optimized for easy surgical handling, tailored degradation to

match the physiological cascade, selective bioactivity to control cellular behavior, and

tissue adhesiveness for facile application. This work resulted in an initial proof-of-concept

device that was shown to effectively prevent adhesion formation in vivo. Future studies

will evaluate the effect of improved retention at the injury site on reducing adhesions and

the benefit of the gelatin matrix in improving anastomotic healing. Given the prevalence

of these procedures and the high morbidity and healthcare costs associated with

complications, the proposed bilayer mesh could have a strong clinical impact.

Page 180: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

157

References

[1] F. Ghaffarpasand, M. Shahrezaei, M. Dehghankhalili, Effects of Platelet Rich Plasma

on Healing Rate of Long Bone Non-union Fractures: A Randomized Double-Blind

Placebo Controlled Clinical Trial, Bull Emerg Trauma 4(3) (2016) 134-140.

[2] P.R. Stafford, B.L. Norris, Reamer-irrigator-aspirator bone graft and bi Masquelet

technique for segmental bone defect nonunions: a review of 25 cases, Injury 41

(2010) S72-S77.

[3] A.C. Masquelet, T. Begue, The Concept of Induced Membrane for Reconstruction of

Long Bone Defects, Orthopedic Clinics 41(1) (2010) 27-37.

[4] P.V. Giannoudis, Treatment of bone defects: Bone transport or the induced membrane

technique?, Injury 47(2) (2016) 291-292.

[5] P. Yin, Q. Ji, T. Li, J. Li, Z. Li, J. Liu, G. Wang, S. Wang, L. Zhang, Z. Mao, P. Tang,

A Systematic Review and Meta-Analysis of Ilizarov Methods in the Treatment of

Infected Nonunion of Tibia and Femur, PLOS ONE 10(11) (2015) e0141973.

[6] R. Morris, M. Hossain, A. Evans, I. Pallister, Induced membrane technique for treating

tibial defects gives mixed results, The Bone & Joint Journal 99-B(5) (2017) 680-

685.

[7] G.I. TAYLOR, G.D.H. MILLER, F.J. HAM, The Free Vascularized Bone Graft: A

Clinical Extension of Microvascular Techniques, Plastic and Reconstructive

Surgery 55(5) (1975) 533-544.

[8] W.C. Pederson, D.W. Person, Long bone reconstruction with vascularized bone grafts,

Orthopedic Clinics 38(1) (2007) 23-35.

[9] M.A. Yee, M.P. Mead, A.I. Alford, D.J. Hak, C. Mauffrey, M.E. Hake, Scientific

understanding of the induced membrane technique: current status and future

directions, Journal of orthopaedic trauma 31 (2017) S3-S8.

[10] C. Mauffrey, M.E. Hake, V. Chadayammuri, A.-C. Masquelet, Reconstruction of

Long Bone Infections Using the Induced Membrane Technique: Tips and Tricks,

Journal of Orthopaedic Trauma 30(6) (2016) e188-e193.

[11] J.M. Anderson, A. Rodriguez, D.T. Chang, Foreign body reaction to biomaterials,

Semin Immunol 20(2) (2008) 86-100.

[12] G. Cox, E. Jones, D. McGonagle, P.V. Giannoudis, Reamer-irrigator-aspirator

indications and clinical results: a systematic review, Int Orthop 35(7) (2011) 951-

956.

[13] R. Gouron, L. Petit, C. Boudot, I. Six, M. Brazier, S. Kamel, R. Mentaverri,

Osteoclasts and their precursors are present in the induced-membrane during bone

Page 181: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

158

reconstruction using the Masquelet technique, Journal of Tissue Engineering and

Regenerative Medicine 11(2) (2017) 382-389.

[14] C. Christou, R.A. Oliver, Y. Yu, W.R. Walsh, The Masquelet technique for membrane

induction and the healing of ovine critical sized segmental defects, PloS one 9(12)

(2014) e114122-e114122.

[15] D. Henrich, C. Seebach, C. Nau, S. Basan, B. Relja, K. Wilhelm, A. Schaible, J. Frank,

J. Barker, I. Marzi, Establishment and characterization of the Masquelet induced

membrane technique in a rat femur critical-sized defect model, Journal of Tissue

Engineering and Regenerative Medicine 10(10) (2016) E382-E396.

[16] X. Wang, F. Wei, F. Luo, K. Huang, Z. Xie, Induction of granulation tissue for the

secretion of growth factors and the promotion of bone defect repair, Journal of

Orthopaedic Surgery and Research 10(1) (2015) 147.

[17] E. Roddy, M.R. DeBaun, A. Daoud-Gray, Y.P. Yang, M.J. Gardner, Treatment of

critical-sized bone defects: clinical and tissue engineering perspectives, European

Journal of Orthopaedic Surgery & Traumatology 28(3) (2018) 351-362.

[18] B.J. Allsopp, D.J. Hunter-Smith, W.M. Rozen, Vascularized versus Nonvascularized

Bone Grafts: What Is the Evidence?, Clin Orthop Relat Res 474(5) (2016) 1319-

1327.

[19] R.J. Cuthbert, S.M. Churchman, H.B. Tan, D. McGonagle, E. Jones, P.V. Giannoudis,

Induced periosteum a complex cellular scaffold for the treatment of large bone

defects, Bone 57(2) (2013) 484-492.

[20] J. Patel, K.P. Gudehithlu, G. Dunea, J.A. Arruda, A.K. Singh, Foreign body-induced

granulation tissue is a source of adult stem cells, Translational Research 155(4)

(2010) 191-199.

[21] P.H. Pelissier, A.C. Masquelet, R. Bareille, S.M. Pelissier, J. Amedee, Induced

membranes secrete growth factors including vascular and osteoinductive factors

and could stimulate bone regeneration, Journal of Orthopaedic Research 22(1)

(2004) 73-79.

[22] Q. Tang, H. Jin, M. Tong, G. Zheng, Z. Xie, S. Tang, J. Jin, P. Shang, H. Xu, L. Shen,

Inhibition of Dll4/Notch1 pathway promotes angiogenesis of Masquelet’s induced

membrane in rats, Experimental & molecular medicine 50(4) (2018) 1-15.

[23] O.-M. Aho, P. Lehenkari, J. Ristiniemi, S. Lehtonen, J. Risteli, H.-V. Leskelä, The

Mechanism of Action of Induced Membranes in Bone Repair, JBJS 95(7) (2013)

597-604.

[24] J. Filipowska, K.A. Tomaszewski, Ł. Niedźwiedzki, J.A. Walocha, T. Niedźwiedzki,

The role of vasculature in bone development, regeneration and proper systemic

functioning, Angiogenesis 20(3) (2017) 291-302.

Page 182: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

159

[25] M. Tarchala, E.J. Harvey, J. Barralet, Biomaterial‐stabilized soft tissue healing for

healing of critical‐sized bone defects: the Masquelet technique, Advanced

healthcare materials 5(6) (2016) 630-640.

[26] A.C. Masquelet, Induced Membrane Technique: Pearls and Pitfalls, Journal of

Orthopaedic Trauma 31 (2017) S36-S38.

[27] P.V. Giannoudis, O. Faour, T. Goff, N. Kanakaris, R. Dimitriou, Masquelet technique

for the treatment of bone defects: tips-tricks and future directions, Injury 42(6)

(2011) 591-598.

[28] V. Chadayammuri, M. Hake, C. Mauffrey, Innovative strategies for the management

of long bone infection: a review of the Masquelet technique, Patient Safety in

Surgery 9(1) (2015) 32.

[29] A. Masquelet, N.K. Kanakaris, L. Obert, P. Stafford, P.V. Giannoudis, Bone Repair

Using the Masquelet Technique, JBJS 101(11) (2019) 1024-1036.

[30] J.-C. Aurégan, T. Bégué, Induced membrane for treatment of critical sized bone

defect: a review of experimental and clinical experiences, Int Orthop 38(9) (2014)

1971-1978.

[31] Y.-f. Ma, N. Jiang, X. Zhang, C.-h. Qin, L. Wang, Y.-j. Hu, Q.-r. Lin, B. Yu, B.-w.

Wang, Calcium sulfate induced versus PMMA-induced membrane in a critical-

sized femoral defect in a rat model, Scientific Reports 8(1) (2018) 637.

[32] N. Gaio, A. Martino, Z. Toth, J.T. Watson, D. Nicolaou, S. McBride-Gagyi, Masquelet

technique: The effect of altering implant material and topography on membrane

matrix composition, mechanical and barrier properties in a rat defect model, J

Biomech 72 (2018) 53-62.

[33] S. McBride‐Gagyi, Z. Toth, D. Kim, V. Ip, E. Evans, J.T. Watson, D. Nicolaou,

Altering spacer material affects bone regeneration in the Masquelet technique in a

rat femoral defect, Journal of Orthopaedic Research® 36(8) (2018) 2228-2238.

[34] V. Luangphakdy, G. Elizabeth Pluhar, N.S. Piuzzi, J.-C. D'Alleyrand, C.S. Carlson,

J.E. Bechtold, J. Forsberg, G.F. Muschler, The Effect of Surgical Technique and

Spacer Texture on Bone Regeneration: A Caprine Study Using the Masquelet

Technique, Clin Orthop Relat Res 475(10) (2017) 2575-2585.

[35] Z. Toth, M. Roi, E. Evans, J.T. Watson, D. Nicolaou, S. McBride-Gagyi, Masquelet

Technique: Effects of Spacer Material and Micro-topography on Factor Expression

and Bone Regeneration, Annals of Biomedical Engineering 47(1) (2019) 174-189.

[36] S.R. Shah, B.T. Smith, A.M. Tatara, E.R. Molina, E.J. Lee, T.C. Piepergerdes, B.A.

Uhrig, R.E. Guldberg, G.N. Bennett, J.C. Wenke, A.G. Mikos, Effects of Local

Antibiotic Delivery from Porous Space Maintainers on Infection Clearance and

Induction of an Osteogenic Membrane in an Infected Bone Defect, Tissue

Engineering Part A 23(3-4) (2017) 91-100.

Page 183: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

160

[37] S.H. McBride, S.F. Evans, M.L. Knothe Tate, Anisotropic mechanical properties of

ovine femoral periosteum and the effects of cryopreservation, J Biomech 44(10)

(2011) 1954-1959.

[38] L.A. Smith, X. Liu, P.X. Ma, Tissue Engineering with Nano-Fibrous Scaffolds, Soft

Matter 4(11) (2008) 2144-2149.

[39] K.M. Woo, V.J. Chen, P.X. Ma, Nano-fibrous scaffolding architecture selectively

enhances protein adsorption contributing to cell attachment, Journal of Biomedical

Materials Research Part A 67A(2) (2003) 531-537.

[40] B.P. Chan, K.W. Leong, Scaffolding in tissue engineering: general approaches and

tissue-specific considerations, European Spine Journal 17(4) (2008) 467-479.

[41] L.A. Smith, P.X. Ma, Nano-fibrous scaffolds for tissue engineering, Colloids and

Surfaces B: Biointerfaces 39(3) (2004) 125-131.

[42] D. Verma, K.S. Katti, D.R. Katti, Polyelectrolyte-complex nanostructured fibrous

scaffolds for tissue engineering, Materials Science and Engineering: C 29(7) (2009)

2079-2084.

[43] H.-F. Ko, C. Sfeir, P.N. Kumta, Novel synthesis strategies for natural polymer and

composite biomaterials as potential scaffolds for tissue engineering, Philosophical

Transactions of the Royal Society A: Mathematical, Physical and Engineering

Sciences 368(1917) (2010) 1981-1997.

[44] J. Koivurinta, E. Hämäläinen, M. Kellomäki, The effect of cross-linking time on a

porous freeze-dried collagen scaffold using 1-ethyl-3-(3-dimethylaminopropyl)

carbodiimide as a cross-linker, Journal of Applied Biomaterials and Biomechanics

6(2) (2008) 89-94.

[45] J. Cheng, Y. Jun, J. Qin, S.-H. Lee, Electrospinning versus microfluidic spinning of

functional fibers for biomedical applications, Biomaterials 114 (2017) 121-143.

[46] H. Hiramatsu, A. Hori, Y. Yajima, M. Yamada, M. Seki, Microfluidics-based wet

spinning of protein microfibers as solid scaffolds for 3D cell cultivation, 2016

International Symposium on Micro-NanoMechatronics and Human Science

(MHS), IEEE, 2016, pp. 1-4.

[47] X. Shi, S. Ostrovidov, Y. Zhao, X. Liang, M. Kasuya, K. Kurihara, K. Nakajima, H.

Bae, H. Wu, A. Khademhosseini, Microfluidic spinning of cell‐responsive grooved

microfibers, Advanced Functional Materials 25(15) (2015) 2250-2259.

[48] K. Wang, X. Chen, Y. Pan, Y. Cui, X. Zhou, D. Kong, Q. Zhao, Enhanced

vascularization in hybrid PCL/gelatin fibrous scaffolds with sustained release of

VEGF, Biomed Res Int 2015 (2015) 865076-865076.

[49] P. Lee, R. Lin, J. Moon, L.P. Lee, Microfluidic alignment of collagen fibers for in

vitro cell culture, Biomedical microdevices 8(1) (2006) 35-41.

Page 184: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

161

[50] C. Haynl, E. Hofmann, K. Pawar, S. Förster, T. Scheibel, Microfluidics-Produced

Collagen Fibers Show Extraordinary Mechanical Properties, Nano Letters 16(9)

(2016) 5917-5922.

[51] S. Soliman, S. Sant, J.W. Nichol, M. Khabiry, E. Traversa, A. Khademhosseini,

Controlling the porosity of fibrous scaffolds by modulating the fiber diameter and

packing density, Journal of Biomedical Materials Research Part A 96A(3) (2011)

566-574.

[52] M. Erencia, F. Cano, J.A. Tornero, M.M. Fernandes, T. Tzanov, J. Macanás, F.

Carrillo, Electrospinning of gelatin fibers using solutions with low acetic acid

concentration: Effect of solvent composition on both diameter of electrospun fibers

and cytotoxicity, Journal of Applied Polymer Science 132(25) (2015).

[53] Z. Ghassemi, G. Slaughter, Cross-linked electrospun gelatin nanofibers for cell-based

assays, Conf Proc IEEE Eng Med Biol Soc 2018 (2018) 6088-6091.

[54] A.P. Kishan, E.M. Cosgriff‐Hernandez, Recent advancements in electrospinning

design for tissue engineering applications: a review, Journal of Biomedical

Materials Research Part A 105(10) (2017) 2892-2905.

[55] Z.S. Patel, S. Young, Y. Tabata, J.A. Jansen, M.E.K. Wong, A.G. Mikos, Dual

delivery of an angiogenic and an osteogenic growth factor for bone regeneration in

a critical size defect model, Bone 43(5) (2008) 931-940.

[56] J. Delgado, E. Sánchez, M. Baro, R. Reyes, C. Évora, A. Delgado, A platelet derived

growth factor delivery system for bone regeneration, Journal of Materials Science:

Materials in Medicine 23(8) (2012) 1903-1912.

[57] S. Bhatia, Natural Polymers vs Synthetic Polymer, Natural Polymer Drug Delivery

Systems: Nanoparticles, Plants, and Algae, Springer International Publishing,

Cham, 2016, pp. 95-118.

[58] L.M. Caballero Aguilar, S.M. Silva, S.E. Moulton, Growth factor delivery: Defining

the next generation platforms for tissue engineering, Journal of Controlled Release

306 (2019) 40-58.

[59] S. Zhang, H. Uludağ, Nanoparticulate systems for growth factor delivery,

Pharmaceutical Research 26(7) (2009) 1561.

[60] Z. Wang, Z. Wang, W.W. Lu, W. Zhen, D. Yang, S. Peng, Novel biomaterial strategies

for controlled growth factor delivery for biomedical applications, NPG Asia

Materials 9(10) (2017) e435-e435.

[61] A.K. Bajpai, S.K. Shukla, S. Bhanu, S. Kankane, Responsive polymers in controlled

drug delivery, Progress in Polymer Science 33(11) (2008) 1088-1118.

[62] M.E. Hoque, T. Nuge, K.Y. Tshai, N. Nordin, V. Prasad, Gelatin Based Scaffolds For

Tissue Engineering – A review, Polymers Research Journal 9 (2015) 15-32.

Page 185: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

162

[63] M.S. Rehmann, K.M. Skeens, P.M. Kharkar, E.M. Ford, E. Maverakis, K.H. Lee,

A.M. Kloxin, Tuning and Predicting Mesh Size and Protein Release from Step

Growth Hydrogels, Biomacromolecules 18(10) (2017) 3131-3142.

[64] C. Addi, F. Murschel, B. Liberelle, N. Riahi, G. De Crescenzo, A highly versatile

adaptor protein for the tethering of growth factors to gelatin-based biomaterials,

Acta Biomaterialia 50 (2017) 198-206.

[65] J.-h. Lee, Y.J. Lee, H.-j. Cho, D.W. Kim, H. Shin, The incorporation of bFGF

mediated by heparin into PCL/gelatin composite fiber meshes for guided bone

regeneration, Drug delivery and translational research 5(2) (2015) 146-159.

[66] X. Zhang, M.R. Battig, N. Chen, E.R. Gaddes, K.L. Duncan, Y. Wang, Chimeric

aptamer–gelatin hydrogels as an extracellular matrix mimic for loading cells and

growth factors, Biomacromolecules 17(3) (2016) 778-787.

[67] S. Basu, S. Pacelli, J. Wang, A. Paul, Adoption of nanodiamonds as biomedical

materials for bone repair, Nanomedicine 12(24) (2017) 2709-2713.

[68] A.K. Gaharwar, L.M. Cross, C.W. Peak, K. Gold, J.K. Carrow, A. Brokesh, K.A.

Singh, 2D Nanoclay for Biomedical Applications: Regenerative Medicine,

Therapeutic Delivery, and Additive Manufacturing, Advanced Materials 31(23)

(2019) 1900332.

[69] G. Cidonio, C.R. Alcala-Orozco, K.S. Lim, M. Glinka, I. Mutreja, Y.-H. Kim, J.I.

Dawson, T.B.F. Woodfield, R.O.C. Oreffo, Osteogenic and angiogenic tissue

formation in high fidelity nanocomposite Laponite-gelatin bioinks, Biofabrication

11(3) (2019) 035027.

[70] J.R. García, A.J. García, Biomaterial-mediated strategies targeting vascularization for

bone repair, Drug delivery and translational research 6(2) (2016) 77-95.

[71] R.J. Bodnar, Chemokine Regulation of Angiogenesis During Wound Healing, Adv

Wound Care (New Rochelle) 4(11) (2015) 641-650.

[72] K.D. Hankenson, M. Dishowitz, C. Gray, M. Schenker, Angiogenesis in bone

regeneration, Injury 42(6) (2011) 556-561.

[73] Y. Bai, G. Yin, Z. Huang, X. Liao, X. Chen, Y. Yao, X. Pu, Localized delivery of

growth factors for angiogenesis and bone formation in tissue engineering,

International Immunopharmacology 16(2) (2013) 214-223.

[74] S. Stegen, N. van Gastel, G. Carmeliet, Bringing new life to damaged bone: The

importance of angiogenesis in bone repair and regeneration, Bone 70 (2015) 19-27.

[75] E. Fagiani, G. Christofori, Angiopoietins in angiogenesis, Cancer Letters 328(1)

(2013) 18-26.

[76] M. Malecki, P. Kolsut, R. Proczka, Angiogenic and antiangiogenic gene therapy, Gene

Therapy 12(1) (2005) S159-S169.

Page 186: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

163

[77] R. Gupta, J. Tongers, D.W. Losordo, Human studies of angiogenic gene therapy, Circ

Res 105(8) (2009) 724-736.

[78] H.-J. Park, F. Yang, S.-W. Cho, Nonviral delivery of genetic medicine for therapeutic

angiogenesis, Advanced Drug Delivery Reviews 64(1) (2012) 40-52.

[79] H. Kasahara, E. Tanaka, N. Fukuyama, E. Sato, H. Sakamoto, Y. Tabata, K. Ando, H.

Iseki, Y. Shinozaki, K. Kimura, E. Kuwabara, S. Koide, H. Nakazawa, H. Mori,

Biodegradable gelatin hydrogel potentiates the angiogenic effect of fibroblast

growth factor 4 plasmid in rabbit hindlimb ischemia, Journal of the American

College of Cardiology 41(6) (2003) 1056-1062.

[80] D. Trentin, H. Hall, S. Wechsler, J.A. Hubbell, Peptide-matrix-mediated gene transfer

of an oxygen-insensitive hypoxia-inducible factor-1α variant for local induction of

angiogenesis, Proceedings of the National Academy of Sciences 103(8) (2006)

2506-2511.

[81] A. Paul, A. Hasan, H.A. Kindi, A.K. Gaharwar, V.T. Rao, M. Nikkhah, S.R. Shin, D.

Krafft, M.R. Dokmeci, D. Shum-Tim, Injectable graphene oxide/hydrogel-based

angiogenic gene delivery system for vasculogenesis and cardiac repair, ACS nano

8(8) (2014) 8050-8062.

[82] J. Kim, A.C. Mirando, A.S. Popel, J.J. Green, Gene delivery nanoparticles to modulate

angiogenesis, Advanced Drug Delivery Reviews 119 (2017) 20-43.

[83] S.-W. Kang, H.-W. Lim, S.-W. Seo, O. Jeon, M. Lee, B.-S. Kim, Nanosphere-

mediated delivery of vascular endothelial growth factor gene for therapeutic

angiogenesis in mouse ischemic limbs, Biomaterials 29(8) (2008) 1109-1117.

[84] J.-H. Jang, C.B. Rives, L.D. Shea, Plasmid Delivery in Vivo from Porous Tissue-

Engineering Scaffolds: Transgene Expression and Cellular Transfection, Molecular

Therapy 12(3) (2005) 475-483.

[85] S.-o. Han, R.I. Mahato, Y.K. Sung, S.W. Kim, Development of Biomaterials for Gene

Therapy, Molecular Therapy 2(4) (2000) 302-317.

[86] K.R. Turner, C. Adams, S. Staelens, H. Deckmyn, J. San Antonio, Crucial Role for

Endothelial Cell α2β1 Integrin Receptor Clustering in Collagen-Induced

Angiogenesis, The Anatomical Record 303(6) (2020) 1604-1618.

[87] A. Duro-Castano, E. Gallon, C. Decker, M.J. Vicent, Modulating angiogenesis with

integrin-targeted nanomedicines, Advanced Drug Delivery Reviews 119 (2017)

101-119.

[88] L.A. Rocha, D.A. Learmonth, R.A. Sousa, A.J. Salgado, αvβ3 and α5β1 integrin-

specific ligands: From tumor angiogenesis inhibitors to vascularization promoters

in regenerative medicine?, Biotechnology Advances 36(1) (2018) 208-227.

Page 187: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

164

[89] J.R. García, A.Y. Clark, A.J. García, Integrin-specific hydrogels functionalized with

VEGF for vascularization and bone regeneration of critical-size bone defects,

Journal of Biomedical Materials Research Part A 104(4) (2016) 889-900.

[90] D.R. Senger, C.A. Perruzzi, M. Streit, V.E. Koteliansky, A.R. de Fougerolles, M.

Detmar, The α1β1 and α2β1 integrins provide critical support for vascular

endothelial growth factor signaling, endothelial cell migration, and tumor

angiogenesis, The American journal of pathology 160(1) (2002) 195-204.

[91] C. Ferris, L. Stevens, K.J. Gilmore, E. Mume, I. Greguric, D. Kirchmajer, G.G.

Wallace, Peptide modification of purified gellan gum, Journal of Materials

Chemistry B 3(6) (2015) 1106-1115.

[92] M.Ø. Dalheim, J. Vanacker, M.A. Najmi, F.L. Aachmann, B.L. Strand, B.E.

Christensen, Efficient functionalization of alginate biomaterials, Biomaterials 80

(2016) 146-156.

[93] E.D. Gomes, S.S. Mendes, H. Leite-Almeida, J.M. Gimble, R.Y. Tam, M.S. Shoichet,

N. Sousa, N.A. Silva, A.J. Salgado, Combination of a peptide-modified gellan gum

hydrogel with cell therapy in a lumbar spinal cord injury animal model,

Biomaterials 105 (2016) 38-51.

[94] Y. Maeshima, U.L. Yerramalla, M. Dhanabal, K.A. Holthaus, S. Barbashov, S.

Kharbanda, C. Reimer, M. Manfredi, W.M. Dickerson, R. Kalluri, Extracellular

matrix-derived peptide binds to αvβ3 integrin and inhibits angiogenesis, Journal of

Biological Chemistry 276(34) (2001) 31959-31968.

[95] M. Nikkhah, N. Eshak, P. Zorlutuna, N. Annabi, M. Castello, K. Kim, A. Dolatshahi-

Pirouz, F. Edalat, H. Bae, Y. Yang, A. Khademhosseini, Directed endothelial cell

morphogenesis in micropatterned gelatin methacrylate hydrogels, Biomaterials

33(35) (2012) 9009-9018.

[96] S.K. Seidlits, C.T. Drinnan, R.R. Petersen, J.B. Shear, L.J. Suggs, C.E. Schmidt,

Fibronectin–hyaluronic acid composite hydrogels for three-dimensional

endothelial cell culture, Acta Biomaterialia 7(6) (2011) 2401-2409.

[97] A.C. Gaffey, M.H. Chen, C.M. Venkataraman, A. Trubelja, C.B. Rodell, P.V. Dinh,

G. Hung, J.W. MacArthur, R.V. Soopan, J.A. Burdick, P. Atluri, Injectable shear-

thinning hydrogels used to deliver endothelial progenitor cells, enhance cell

engraftment, and improve ischemic myocardium, The Journal of Thoracic and

Cardiovascular Surgery 150(5) (2015) 1268-1277.

[98] X. Wu, E. Rabkin-Aikawa, K.J. Guleserian, T.E. Perry, Y. Masuda, F.W.H.

Sutherland, F.J. Schoen, J. John E. Mayer, J. Bischoff, Tissue-engineered

microvessels on three-dimensional biodegradable scaffolds using human

endothelial progenitor cells, American Journal of Physiology-Heart and Circulatory

Physiology 287(2) (2004) H480-H487.

Page 188: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

165

[99] G. Camci-Unal, N. Alemdar, N. Annabi, A. Khademhosseini, Oxygen-releasing

biomaterials for tissue engineering, Polymer International 62(6) (2013) 843-848.

[100] B.S. Harrison, D. Eberli, S.J. Lee, A. Atala, J.J. Yoo, Oxygen producing biomaterials

for tissue regeneration, Biomaterials 28(31) (2007) 4628-4634.

[101] S.H. Oh, C.L. Ward, A. Atala, J.J. Yoo, B.S. Harrison, Oxygen generating scaffolds

for enhancing engineered tissue survival, Biomaterials 30(5) (2009) 757-762.

[102] S.-M. Ng, J.-Y. Choi, H.-S. Han, J.-S. Huh, J.O. Lim, Novel microencapsulation of

potential drugs with low molecular weight and high hydrophilicity: Hydrogen

peroxide as a candidate compound, International Journal of Pharmaceutics 384(1)

(2010) 120-127.

[103] B. Taylor, B. French, T. Fowler, J. Russell, A. Poka, Induced Membrane Technique

for Reconstruction To Manage Bone Loss, The Journal of the American Academy

of Orthopaedic Surgeons 20 (2012) 142-50.

[104] M.L. Azi, A.A.d.A. Teixeira, R.B. Cotias, A. Joeris, M.J. Kfuri, Membrane Induced

Osteogenesis in the Management of Posttraumatic Bone Defects, Journal of

Orthopaedic Trauma 30(10) (2016) 545-550.

[105] T. Apard, N. Bigorre, P. Cronier, F. Duteille, P. Bizot, P. Massin, Two-stage

reconstruction of post-traumatic segmental tibia bone loss with nailing,

Orthopaedics & Traumatology: Surgery & Research 96(5) (2010) 549-553.

[106] M. Angel, C.P.-F. Juan, R.-R. Carmen, S.-G. Carmen, H.A. Mohammad, R. Rosa,

R.-G. Arantza, B. Ana, L.-C. Enrique, Antibiotic Release from Calcium Phosphate

Materials in Oral and Maxillofacial Surgery. Molecular, Cellular and

Pharmaceutical Aspects, Current Pharmaceutical Biotechnology 18(1) (2017) 52-

63.

[107] A.D. Hanssen, Local Antibiotic Delivery Vehicles in the Treatment of

Musculoskeletal Infection, Clinical Orthopaedics and Related Research® 437

(2005) 91-96.

[108] N.W. Cortés-Penfield, P.A. Kulkarni, The history of antibiotic treatment of

osteomyelitis, Open forum infectious diseases, Oxford University Press US, 2019,

p. ofz181.

[109] G. Kapoor, S. Saigal, A. Elongavan, Action and resistance mechanisms of

antibiotics: A guide for clinicians, Journal of anaesthesiology, clinical

pharmacology 33(3) (2017) 300-305.

[110] G. Kaufman, Antibiotics: mode of action and mechanisms of resistance, Nursing

standard 25(42) (2011).

[111] C.J. Mathews, Bone and joint infections, Medicine 42(5) (2014) 266-270.

Page 189: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

166

[112] H.S. Fraimow, Systemic antimicrobial therapy in osteomyelitis, Semin Plast Surg,

Thieme Medical Publishers, 2009, p. 90.

[113] L.S. Gonzalez III, J.P. Spencer, Aminoglycosides: a practical review, American

family physician 58(8) (1998) 1811.

[114] F. Dowd, B. Johnson, A. Mariotti, P. Kumar, Pharmacology of specific drug groups:

antibiotic therapy, Pharmacol. Ther. Dent (2017) 457-487.

[115] K.F. KONG, L. Schneper, K. Mathee, Beta‐lactam antibiotics: from antibiosis to

resistance and bacteriology, Apmis 118(1) (2010) 1-36.

[116] J. Popovich, M. Stephens, H. Celaya, S. Suwarno, S. Barclay, E. Yee, D.A. Dean,

M. Farris, S.E. Haydel, Building and Breaking the Cell Wall in Four Acts: A

Kinesthetic and Tactile Role-Playing Exercise for Teaching Beta-Lactam

Antibiotic Mechanism of Action and Resistance, Journal of microbiology &

biology education 19(1) (2018).

[117] J.M. Blondeau, Fluoroquinolones: mechanism of action, classification, and

development of resistance, Survey of ophthalmology 49(2) (2004) S73-S78.

[118] D. Zeng, D. Debabov, T.L. Hartsell, R.J. Cano, S. Adams, J.A. Schuyler, R.

McMillan, J.L. Pace, Approved glycopeptide antibacterial drugs: mechanism of

action and resistance, Cold Spring Harbor perspectives in medicine 6(12) (2016)

a026989.

[119] G.R. Hartmann, P. Heinrich, M.C. Kollenda, B. Skrobranek, M. Tropschug, W.

Weiß, Molecular mechanism of action of the antibiotic rifampicin, Angewandte

Chemie International Edition in English 24(12) (1985) 1009-1014.

[120] I. Chopra, M. Roberts, Tetracycline antibiotics: mode of action, applications,

molecular biology, and epidemiology of bacterial resistance, Microbiology and

molecular biology reviews 65(2) (2001) 232-260.

[121] B. Spellberg, B.A. Lipsky, Systemic antibiotic therapy for chronic osteomyelitis in

adults, Clinical infectious diseases 54(3) (2012) 393-407.

[122] P. Zhang, J. Qin, B. Zhang, Y. Zheng, L. Yang, Y. Shen, B. Zuo, F. Zhang,

Gentamicin-loaded silk/nanosilver composite scaffolds for MRSA-induced chronic

osteomyelitis, Royal Society open science 6(5) (2019) 182102.

[123] J.S. Moskowitz, M.R. Blaisse, R.E. Samuel, H.-P. Hsu, M.B. Harris, S.D. Martin,

J.C. Lee, M. Spector, P.T. Hammond, The effectiveness of the controlled release of

gentamicin from polyelectrolyte multilayers in the treatment of Staphylococcus

aureus infection in a rabbit bone model, Biomaterials 31(23) (2010) 6019-6030.

[124] C. Jacqueline, G. Amador, J. Caillon, V. Le Mabecque, E. Batard, A.-F. Miègeville,

D. Biek, Y. Ge, G. Potel, A. Hamel, Efficacy of the new cephalosporin ceftaroline

in the treatment of experimental methicillin-resistant Staphylococcus aureus acute

osteomyelitis, Journal of antimicrobial chemotherapy 65(8) (2010) 1749-1752.

Page 190: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

167

[125] A.C. Matos, C.F. Marques, R.V. Pinto, I.A. Ribeiro, L.M. Gonçalves, M.A. Vaz, J.

Ferreira, A.J. Almeida, A.F. Bettencourt, Novel doped calcium phosphate-PMMA

bone cement composites as levofloxacin delivery systems, International journal of

pharmaceutics 490(1-2) (2015) 200-208.

[126] J. Zhou, X. Zhou, J. Wang, H. Zhou, J. Dong, Treatment of osteomyelitis defects by

a vancomycin-loaded gelatin/β-tricalcium phosphate composite scaffold, Bone &

Joint Research 7(1) (2018) 46-57.

[127] P. Vergidis, M.S. Rouse, G. Euba, M.J. Karau, S.M. Schmidt, J.N. Mandrekar, J.M.

Steckelberg, R. Patel, Treatment with linezolid or vancomycin in combination with

rifampin is effective in an animal model of methicillin-resistant Staphylococcus

aureus foreign body osteomyelitis, Antimicrobial agents and chemotherapy 55(3)

(2011) 1182-1186.

[128] D. Stengel, K. Bauwens, J. Sehouli, A. Ekkernkamp, F. Porzsolt, Systematic review

and meta-analysis of antibiotic therapy for bone and joint infections, The Lancet

infectious diseases 1(3) (2001) 175-188.

[129] S.B. Zaman, M.A. Hussain, R. Nye, V. Mehta, K.T. Mamun, N. Hossain, A review

on antibiotic resistance: alarm bells are ringing, Cureus 9(6) (2017).

[130] W. Sim, R.T. Barnard, M. Blaskovich, Z.M. Ziora, Antimicrobial silver in medicinal

and consumer applications: A patent review of the past decade (2007–2017),

Antibiotics 7(4) (2018) 93.

[131] J.A. Lemire, J.J. Harrison, R.J. Turner, Antimicrobial activity of metals:

mechanisms, molecular targets and applications, Nature Reviews Microbiology

11(6) (2013) 371-384.

[132] G.V. Vimbela, S.M. Ngo, C. Fraze, L. Yang, D.A. Stout, Antibacterial properties

and toxicity from metallic nanomaterials, International journal of nanomedicine 12

(2017) 3941.

[133] J.-Y. Maillard, P. Hartemann, Silver as an antimicrobial: facts and gaps in

knowledge, Critical reviews in microbiology 39(4) (2013) 373-383.

[134] R.J. Turner, Metal-based antimicrobial strategies, Microb Biotechnol 10(5) (2017)

1062-1065.

[135] L.C. Yun’an Qing, R. Li, G. Liu, Y. Zhang, X. Tang, J. Wang, H. Liu, Y. Qin,

Potential antibacterial mechanism of silver nanoparticles and the optimization of

orthopedic implants by advanced modification technologies, International journal

of nanomedicine 13 (2018) 3311.

[136] J. Pasquet, Y. Chevalier, J. Pelletier, E. Couval, D. Bouvier, M.-A. Bolzinger, The

contribution of zinc ions to the antimicrobial activity of zinc oxide, Colloids and

Surfaces A: Physicochemical and Engineering Aspects 457 (2014) 263-274.

Page 191: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

168

[137] M.M. Almoudi, A.S. Hussein, M.I.A. Hassan, N.M. Zain, A systematic review on

antibacterial activity of zinc against Streptococcus mutans, The Saudi dental

journal 30(4) (2018) 283-291.

[138] A.B. Kelson, M. Carnevali, V. Truong-Le, Gallium-based anti-infectives: targeting

microbial iron-uptake mechanisms, Current opinion in pharmacology 13(5) (2013)

707-716.

[139] L. Bernstein, Gallium, therapeutic effects, Encyclopedia of metalloproteins (2013)

823-835.

[140] S. Hijazi, D. Visaggio, M. Pirolo, E. Frangipani, L. Bernstein, P. Visca,

Antimicrobial activity of gallium compounds on ESKAPE pathogens, Frontiers in

cellular and infection microbiology 8 (2018) 316.

[141] S. Cereceres, Z. Lan, L. Bryan, M. Whitely, T. Wilems, H. Greer, E.R. Alexander,

R.J. Taylor, L. Bernstein, N. Cohen, C. Whitfield-Cargile, E. Cosgriff-Hernandez,

W.S. C., D.V. R., W.J. S., A.D. G., Bactericidal activity of 3D-printed hydrogel

dressing loaded with gallium maltolate, APL Bioengineering 3(2) (2019) 026102.

[142] S.K. Nandi, A. Shivaram, S. Bose, A. Bandyopadhyay, Silver nanoparticle deposited

implants to treat osteomyelitis, Journal of Biomedical Materials Research Part B:

Applied Biomaterials 106(3) (2018) 1073-1083.

[143] N. Garino, P. Sanvitale, B. Dumontel, M. Laurenti, M. Colilla, I. Izquierdo-Barba,

V. Cauda, M. Vallet-Regì, Zinc oxide nanocrystals as a nanoantibiotic and

osteoinductive agent, RSC advances 9(20) (2019) 11312-11321.

[144] A. Cochis, B. Azzimonti, R. Chiesa, L. Rimondini, M. Gasik, Metallurgical Gallium

Additions to Titanium Alloys Demonstrate a Strong Time-Increasing Antibacterial

Activity without any Cellular Toxicity, ACS Biomaterials Science & Engineering

5(6) (2019) 2815-2820.

[145] N.S. Moghaddam, M.T. Andani, A. Amerinatanzi, C. Haberland, S. Huff, M. Miller,

M. Elahinia, D. Dean, Metals for bone implants: Safety, design, and efficacy,

Biomanufacturing Reviews 1(1) (2016) 1.

[146] N.K. Brogden, K.A. Brogden, Will new generations of modified antimicrobial

peptides improve their potential as pharmaceuticals?, International journal of

antimicrobial agents 38(3) (2011) 217-225.

[147] B. Mishra, S. Reiling, D. Zarena, G. Wang, Host defense antimicrobial peptides as

antibiotics: design and application strategies, Current opinion in chemical biology

38 (2017) 87-96.

[148] S.A. Baltzer, M.H. Brown, Antimicrobial peptides–promising alternatives to

conventional antibiotics, Journal of molecular microbiology and biotechnology

20(4) (2011) 228-235.

Page 192: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

169

[149] B. Bechinger, S.-U. Gorr, Antimicrobial peptides: mechanisms of action and

resistance, Journal of dental research 96(3) (2017) 254-260.

[150] H. Jenssen, P. Hamill, R.E. Hancock, Peptide antimicrobial agents, Clinical

microbiology reviews 19(3) (2006) 491-511.

[151] M.-D. Seo, H.-S. Won, J.-H. Kim, T. Mishig-Ochir, B.-J. Lee, Antimicrobial

peptides for therapeutic applications: a review, Molecules 17(10) (2012) 12276-

12286.

[152] M. Amerikova, I. Pencheva El-Tibi, V. Maslarska, S. Bozhanov, K. Tachkov,

Antimicrobial activity, mechanism of action, and methods for stabilisation of

defensins as new therapeutic agents, Biotechnology & Biotechnological Equipment

33(1) (2019) 671-682.

[153] I. Zelezetsky, A. Tossi, Alpha-helical antimicrobial peptides—Using a sequence

template to guide structure–activity relationship studies, Biochimica et Biophysica

Acta (BBA)-Biomembranes 1758(9) (2006) 1436-1449.

[154] L.T. Nguyen, E.F. Haney, H.J. Vogel, The expanding scope of antimicrobial peptide

structures and their modes of action, Trends in biotechnology 29(9) (2011) 464-

472.

[155] Y. Park, K. Hahm, Antimicrobial peptides (AMPs): peptide structure and mode of

action, Journal of biochemistry and molecular biology 38(5) (2005) 507.

[156] H.P. Stallmann, C. Faber, A.V.N. Amerongen, P.I. Wuisman, Antimicrobial

peptides: review of their application in musculoskeletal infections, Injury 37(2)

(2006) S34-S40.

[157] A.M. Eid, A.A. Eid, Multidrug resistance in bone infection: antimicrobial peptides

as a therapeutic tool, The Egyptian Orthopaedic Journal 48(2) (2013) 113.

[158] M. Kazemzadeh‐Narbat, S. Noordin, B.A. Masri, D.S. Garbuz, C.P. Duncan, R.E.

Hancock, R. Wang, Drug release and bone growth studies of antimicrobial peptide‐

loaded calcium phosphate coating on titanium, Journal of Biomedical Materials

Research Part B: Applied Biomaterials 100(5) (2012) 1344-1352.

[159] P. Melicherčík, O. Nešuta, V. Čeřovský, Antimicrobial peptides for topical treatment

of osteomyelitis and implant-related infections: study in the spongy bone,

Pharmaceuticals 11(1) (2018) 20.

[160] C. Faber, H.P. Stallmann, D. Lyaruu, U. Joosten, C. von Eiff, A. van Nieuw

Amerongen, P.I. Wuisman, Comparable efficacies of the antimicrobial peptide

human lactoferrin 1-11 and gentamicin in a chronic methicillin-resistant

Staphylococcus aureus osteomyelitis model, Antimicrobial agents and

chemotherapy 49(6) (2005) 2438-2444.

Page 193: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

170

[161] J.G.E. Hendriks, J.R. van Horn, H.C. van der Mei, H.J. Busscher, Backgrounds of

antibiotic-loaded bone cement and prosthesis-related infection, Biomaterials 25(3)

(2004) 545-556.

[162] C. Nau, C. Seebach, A. Trumm, A. Schaible, K. Kontradowitz, S. Meier, H.

Buechner, I. Marzi, D. Henrich, Alteration of Masquelet's induced membrane

characteristics by different kinds of antibiotic enriched bone cement in a critical

size defect model in the rat's femur, Injury 47(2) (2016) 325-334.

[163] J.A. Inzana, E.M. Schwarz, S.L. Kates, H.A. Awad, Biomaterials approaches to

treating implant-associated osteomyelitis, Biomaterials 81 (2016) 58-71.

[164] B. Li, K.V. Brown, J.C. Wenke, S.A. Guelcher, Sustained release of vancomycin

from polyurethane scaffolds inhibits infection of bone wounds in a rat femoral

segmental defect model, Journal of Controlled Release 145(3) (2010) 221-230.

[165] O. Kluin, H. van der Mei, H. Busscher, D. Neut, Biodegradable vs non-biodegradable

antibiotic delivery devices in the treatment of osteomyelitis, Expert opinion on drug

delivery 10 (2013).

[166] J. Ferguson, M. Dudareva, N. Riley, D. Stubbs, B. Atkins, M. McNally, The use of

a biodegradable antibiotic-loaded calcium sulphate carrier containing tobramycin

for the treatment of chronic osteomyelitis: a series of 195 cases, The bone & joint

journal 96(6) (2014) 829-836.

[167] P. Gao, X. Nie, M. Zou, Y. Shi, G. Cheng, Recent advances in materials for extended-

release antibiotic delivery system, The Journal of Antibiotics 64(9) (2011) 625-634.

[168] M.A.T. Aroni, P.F.D. Costa Neto, G.J.P.L.D. Oliveira, R.A.C. Marcantonio, E.

Marcantonio Junior, Bone repair induced by different bone graft substitutes in

critical-sized defects in rat calvaria, Revista de Odontologia da UNESP 48 (2019).

[169] P.-F. Wei, Z.-Y. Yuan, W. Jing, B.-B. Guan, Z.-H. Liu, X. Zhang, J.-P. Mao, D.-F.

Chen, Q. Cai, X.-P. Yang, Regenerating infected bone defects with

osteocompatible microspheres possessing antibacterial activity, Biomaterials

science 7(1) (2019) 272-286.

[170] J. Hatzenbuehler, T.J. Pulling, Diagnosis and management of osteomyelitis,

American family physician 84(9) (2011) 1027-1033.

[171] H.M. Nguyen, C.J. Graber, Limitations of antibiotic options for invasive infections

caused by methicillin-resistant Staphylococcus aureus: is combination therapy the

answer?, Journal of Antimicrobial Chemotherapy 65(1) (2009) 24-36.

[172] J.D. Caplin, A.J. García, Implantable antimicrobial biomaterials for local drug

delivery in bone infection models, Acta Biomaterialia 93 (2019) 2-11.

[173] K. Vasilev, J. Cook, H.J. Griesser, Antibacterial surfaces for biomedical devices,

Expert Review of Medical Devices 6(5) (2009) 553-67.

Page 194: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

171

[174] S. Kasha, S.S. Rathore, H. Kumar, Antibiotic Cement Spacer and Induced Membrane

Bone Grafting in Open Fractures with Bone Loss: A Case Series, Indian J Orthop

53(2) (2019) 237-245.

[175] T.M. Wong, T.W. Lau, X. Li, C. Fang, K. Yeung, F. Leung, Masquelet Technique

for Treatment of Posttraumatic Bone Defects, The Scientific World Journal 2014

(2014) 710302.

[176] J.A. Lyndon, B.J. Boyd, N. Birbilis, Metallic implant drug/device combinations for

controlled drug release in orthopaedic applications, Journal of Controlled Release

179 (2014) 63-75.

[177] J. Wang, L. Wang, Z. Zhou, H. Lai, P. Xu, L. Liao, J. Wei, Biodegradable Polymer

Membranes Applied in Guided Bone/Tissue Regeneration: A Review, Polymers

8(4) (2016) 115.

[178] S.P. Noel, H. Courtney, J.D. Bumgardner, W.O. Haggard, Chitosan Films: A

Potential Local Drug Delivery System for Antibiotics, Clin Orthop Relat Res

466(6) (2008) 1377-1382.

[179] S. Jin, J. Li, J. Wang, J. Jiang, Y. Zuo, Y. Li, F. Yang, Electrospun silver ion-loaded

calcium phosphate/chitosan antibacterial composite fibrous membranes for guided

bone regeneration, International journal of nanomedicine 13 (2018) 4591-4605.

[180] S. Radin, P. Ducheyne, Controlled release of vancomycin from thin sol–gel films on

titanium alloy fracture plate material, Biomaterials 28(9) (2007) 1721-1729.

[181] T.S. Sørensen, l.I. Sørensen, S. Merser, Rapid release of gentamicin from collagen

sponge: in vitro comparison with plastic beads, Acta orthopaedica Scandinavica

61(4) (1990) 353-356.

[182] O. Kilian, H. Hossain, I. Flesch, U. Sommer, H. Nolting, T. Chakraborty, R.

Schnettler, Elution kinetics, antimicrobial efficacy, and degradation and

microvasculature of a new gentamicin‐loaded collagen fleece, Journal of

Biomedical Materials Research Part B: Applied Biomaterials 90(1) (2009) 210-

222.

[183] H.F. Chuang, R.C. Smith, P.T. Hammond, Polyelectrolyte Multilayers for Tunable

Release of Antibiotics, Biomacromolecules 9(6) (2008) 1660-1668.

[184] E.A. Masters, R.P. Trombetta, K.L. de Mesy Bentley, B.F. Boyce, A.L. Gill, S.R.

Gill, K. Nishitani, M. Ishikawa, Y. Morita, H. Ito, S.N. Bello-Irizarry, M.

Ninomiya, J.D. Brodell, Jr., C.C. Lee, S.P. Hao, I. Oh, C. Xie, H.A. Awad, J.L.

Daiss, J.R. Owen, S.L. Kates, E.M. Schwarz, G. Muthukrishnan, Evolving concepts

in bone infection: redefining "biofilm", "acute vs. chronic osteomyelitis", "the

immune proteome" and "local antibiotic therapy", Bone Res 7 (2019) 20-20.

Page 195: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

172

[185] M. Aviv, I. Berdicevsky, M. Zilberman, Gentamicin-loaded bioresorbable films for

prevention of bacterial infections associated with orthopedic implants, Journal of

Biomedical Materials Research Part A 83A(1) (2007) 10-19.

[186] X. Zhang, U.P. Wyss, D. Pichora, M.F. Goosen, Biodegradable controlled antibiotic

release devices for osteomyelitis: optimization of release properties, Journal of

pharmacy and pharmacology 46(9) (1994) 718-724.

[187] K. Ye, H. Kuang, Z. You, Y. Morsi, X. Mo, Electrospun Nanofibers for Tissue

Engineering with Drug Loading and Release, Pharmaceutics 11(4) (2019) 182.

[188] J. Gao, G. Huang, G. Liu, Y. Liu, Q. Chen, L. Ren, C. Chen, Z. Ding, A

biodegradable antibiotic-eluting PLGA nanofiber-loaded deproteinized bone for

treatment of infected rabbit bone defects, Journal of Biomaterials Applications

31(2) (2016) 241-249.

[189] S. Wei, C. Jian, F. Xu, T. Bao, S. Lan, G. Wu, B. Qi, Z. Bai, A. Yu, Vancomycin–

impregnated electrospun polycaprolactone (PCL) membrane for the treatment of

infected bone defects: An animal study, Journal of Biomaterials Applications 32(9)

(2018) 1187-1196.

[190] H. Saderi, P. OULIA, N.M. JALALI, Difference in epidemiology and antibiotic

susceptibility of methicillin resistant and methicillin susceptible Staphylococcus

aureus isolates, (2009).

[191] M. Coleman, K. Kuskie, M. Liu, K. Chaffin, M. Libal, S. Giguère, L. Bernstein, N.

Cohen, In vitro antimicrobial activity of gallium maltolate against virulent

Rhodococcus equi, Veterinary Microbiology 146(1) (2010) 175-178.

[192] L. Giacani, L.R. Bernstein, A.M. Haynes, B.C. Godornes, G. Ciccarese, F. Drago,

A. Parodi, S. Valdevit, L. Anselmi, C.F. Tomasini, A.M. Baca, Topical treatment

with gallium maltolate reduces Treponema pallidum subsp. pertenue burden in

primary experimental lesions in a rabbit model of yaws, PLOS Neglected Tropical

Diseases 13(1) (2019) e0007076.

[193] P. Frutos, S. Torrado, M.E. Perez-Lorenzo, G. Frutos, A validated quantitative

colorimetric assay for gentamicin, Journal of Pharmaceutical and Biomedical

Analysis 21(6) (2000) 1149-1159.

[194] J. Hudzicki, Kirby-Bauer disk diffusion susceptibility test protocol, (2009).

[195] V.H. Tam, S. Kabbara, G. Vo, A.N. Schilling, E.A. Coyle, Comparative

pharmacodynamics of gentamicin against Staphylococcus aureus and

Pseudomonas aeruginosa, Antimicrobial agents and chemotherapy 50(8) (2006)

2626-2631.

[196] C.C. Llanos-Paez, S. Hennig, C.E. Staatz, Population pharmacokinetic modelling,

Monte Carlo simulation and semi-mechanistic pharmacodynamic modelling as

Page 196: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

173

tools to personalize gentamicin therapy, Journal of Antimicrobial Chemotherapy

72(3) (2017) 639-667.

[197] K. Kanellakopoulou, E.J. Giamarellos-Bourboulis, Carrier Systems for the Local

Delivery of Antibiotics in Bone Infections, Drugs 59(6) (2000) 1223-1232.

[198] C.L.S. Institute, Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria

That Grow Aerobically; Approved Standards—Ninth Edition, 2012.

[199] D. Baldoni, A. Steinhuber, W. Zimmerli, A. Trampuz, In vitro activity of gallium

maltolate against Staphylococci in logarithmic, stationary, and biofilm growth

phases: comparison of conventional and calorimetric susceptibility testing

methods, Antimicrobial agents and chemotherapy 54(1) (2010) 157-163.

[200] K. Kim, Y.K. Luu, C. Chang, D. Fang, B.S. Hsiao, B. Chu, M. Hadjiargyrou,

Incorporation and controlled release of a hydrophilic antibiotic using poly (lactide-

co-glycolide)-based electrospun nanofibrous scaffolds, Journal of controlled

release 98(1) (2004) 47-56.

[201] M. Brewster, G. Verreck, I. Chun, J. Rosenblatt, J. Mensch, A. Van Dijck, M. Noppe,

A. Arien, M. Bruining, J. Peeters, The use of polymer-based electrospun nanofibers

containing amorphous drug dispersions for the delivery of poorly water-soluble

pharmaceuticals, Die Pharmazie-An International Journal of Pharmaceutical

Sciences 59(5) (2004) 387-391.

[202] G. Kapoor, S. Saigal, A. Elongavan, Action and resistance mechanisms of

antibiotics: A guide for clinicians, J Anaesthesiol Clin Pharmacol 33(3) (2017) 300-

305.

[203] W. Chaisri, A.H. Ghassemi, W.E. Hennink, S. Okonogi, Enhanced gentamicin

loading and release of PLGA and PLHMGA microspheres by varying the

formulation parameters, Colloids and Surfaces B: Biointerfaces 84(2) (2011) 508-

514.

[204] P. Coimbra, J.P. Freitas, T. Gonçalves, M.H. Gil, M. Figueiredo, Preparation of

gentamicin sulfate eluting fiber mats by emulsion and by suspension

electrospinning, Materials Science and Engineering: C 94 (2019) 86-93.

[205] H.K. Makadia, S.J. Siegel, Poly Lactic-co-Glycolic Acid (PLGA) as Biodegradable

Controlled Drug Delivery Carrier, Polymers 3(3) (2011) 1377-1397.

[206] A. Rangel-Vega, L. Bernstein, E.-A. Mandujano Tinoco, S.-J. García-Contreras, R.

García-Contreras, Drug repurposing as an alternative for the treatment of

recalcitrant bacterial infections, Frontiers in Microbiology 6(282) (2015).

[207] R. Chandki, P. Banthia, R. Banthia, Biofilms: A microbial home, J Indian Soc

Periodontol 15(2) (2011) 111-114.

Page 197: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

174

[208] J. Penn-Barwell, C. Murray, J. Wenke, Early antibiotics and debridement

independently reduce infection in an open fracture model, The Journal of Bone and

Joint Surgery. British volume 94(1) (2012) 107-112.

[209] M.V. Natu, H.C. de Sousa, M.H. Gil, Effects of drug solubility, state and loading on

controlled release in bicomponent electrospun fibers, International Journal of

Pharmaceutics 397(1) (2010) 50-58.

[210] M. Zamani, M. Morshed, J. Varshosaz, M. Jannesari, Controlled release of

metronidazole benzoate from poly ε-caprolactone electrospun nanofibers for

periodontal diseases, European Journal of Pharmaceutics and Biopharmaceutics

75(2) (2010) 179-185.

[211] H.S. Fraimow, Systemic antimicrobial therapy in osteomyelitis, Semin Plast Surg

23(2) (2009) 90-99.

[212] S.P. Lawless, N.D. Cohen, S.D. Lawhon, A.M. Chamoun-Emanuelli, J. Wu, A.

Rivera-Vélez, B.R. Weeks, C.M. Whitfield-Cargile, Effect of gallium maltolate on

a model of chronic, infected equine distal limb wounds, PloS one 15(6) (2020)

e0235006.

[213] A. Khademhosseini, R. Langer, A decade of progress in tissue engineering, Nature

Protocols 11 (2016) 1775.

[214] J.A. Hubbell, Bioactive biomaterials, Current opinion in biotechnology 10(2) (1999)

123-129.

[215] T.P. Richardson, M.C. Peters, A.B. Ennett, D.J. Mooney, Polymeric system for dual

growth factor delivery, Nature Biotechnology 19 (2001) 1029.

[216] V. Luginbuehl, L. Meinel, H.P. Merkle, B. Gander, Localized delivery of growth

factors for bone repair, European journal of pharmaceutics and biopharmaceutics

58(2) (2004) 197-208.

[217] M.E. Hoque, T. Nuge, T.K. Yeow, N. Nordin, R. Prasad, Gelatin based scaffolds for

tissue engineering-a review, Polymers Research Journal 9(1) (2015) 15.

[218] N. Alemdar, Fabrication of a novel bone ash-reinforced gelatin/alginate/hyaluronic

acid composite film for controlled drug delivery, Carbohydrate Polymers 151

(2016) 1019-1026.

[219] T. Kobayashi, M. Mizuta, N. Hiwatashi, Y. Kishimoto, T. Nakamura, S.-i.

Kanemaru, S. Hirano, Drug delivery system of basic fibroblast growth factor using

gelatin hydrogel for restoration of acute vocal fold scar, Auris Nasus Larynx 44(1)

(2017) 86-92.

[220] N. Hiwatashi, S. Hirano, M. Mizuta, I. Tateya, S.-i. Kanemaru, T. Nakamura, J. Ito,

K. Kawai, S. Suzuki, Biocompatibility and Efficacy of Collagen/Gelatin Sponge

Scaffold With Sustained Release of Basic Fibroblast Growth Factor on Vocal Fold

Page 198: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

175

Fibroblasts in 3-Dimensional Culture, Annals of Otology, Rhinology &

Laryngology 124(2) (2015) 116-125.

[221] P.N. Dang, N. Dwivedi, L.M. Phillips, X. Yu, S. Herberg, C. Bowerman, L.D.

Solorio, W.L. Murphy, E. Alsberg, Controlled dual growth factor delivery from

microparticles incorporated within human bone marrow‐derived mesenchymal

stem cell aggregates for enhanced bone tissue engineering via endochondral

ossification, Stem cells translational medicine 5(2) (2016) 206-217.

[222] F. Hirose, J. Kiryu, Y. Tabata, H. Tamura, K. Musashi, N. Takase, H. Usui, S.

Kuwayama, A. Kato, N. Yoshimura, Y. Ogura, T. Yasukawa, Experimental

proliferative vitreoretinopathy in rabbits by delivery of bioactive proteins with

gelatin microspheres, European Journal of Pharmaceutics and Biopharmaceutics

129 (2018) 267-272.

[223] A. Sionkowska, S. Skrzyński, K. Śmiechowski, A. Kołodziejczak, The review of

versatile application of collagen, Polymers for Advanced Technologies 28(1)

(2017) 4-9.

[224] M. Lalande, C. Comby-Zerbino, M. Bouakil, P. Dugourd, F. Chirot, J.-C. Poully,

Isolated Collagen Mimetic Peptide Assemblies Have Stable Triple-Helix

Structures, Chemistry – A European Journal 24(52) (2018) 13728-13733.

[225] P. Aramwit, N. Jaichawa, J. Ratanavaraporn, T. Srichana, A comparative study of

type A and type B gelatin nanoparticles as the controlled release carriers for

different model compounds, Materials Express 5(3) (2015) 241-248.

[226] M. Foox, M. Zilberman, Drug delivery from gelatin-based systems, Expert Opinion

on Drug Delivery 12(9) (2015) 1547-1563.

[227] M. Nikkhah, Akbari, M. , Paul, A. , Memic, A. , Dolatshahi‐Pirouz, A. and

Khademhosseini, A., Gelatin-Based Biomaterials For Tissue Engineering And

Stem Cell Bioengineering, Biomaterials from Nature for Advanced Devices and

Therapies2016, pp. 37-62.

[228] S. Afewerki, A. Sheikhi, S. Kannan, S. Ahadian, A. Khademhosseini, Gelatin-

polysaccharide composite scaffolds for 3D cell culture and tissue engineering:

Towards natural therapeutics, Bioengineering & Translational Medicine 4(1)

(2019) 96-115.

[229] M. Nikkhah, M. Akbari, A. Paul, A. Memic, A. Dolatshahi‐Pirouz, A.

Khademhosseini, Gelatin‐Based Biomaterials For Tissue Engineering And Stem

Cell Bioengineering, Biomaterials from Nature for Advanced Devices and

Therapies (2016) 37-62.

[230] W.-H. Lin, J. Yu, G. Chen, W.-B. Tsai, Fabrication of multi-biofunctional gelatin-

based electrospun fibrous scaffolds for enhancement of osteogenesis of

mesenchymal stem cells, Colloids and Surfaces B: Biointerfaces 138 (2016) 26-31.

Page 199: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

176

[231] H. Lee, S. Lim, M.S. Birajdar, S.-H. Lee, H. Park, Fabrication of FGF-2 immobilized

electrospun gelatin nanofibers for tissue engineering, International Journal of

Biological Macromolecules 93 (2016) 1559-1566.

[232] J. Ratanavaraporn, R. Rangkupan, H. Jeeratawatchai, S. Kanokpanont, S.

Damrongsakkul, Influences of physical and chemical crosslinking techniques on

electrospun type A and B gelatin fiber mats, International Journal of Biological

Macromolecules 47(4) (2010) 431-438.

[233] C.N. Grover, J.H. Gwynne, N. Pugh, S. Hamaia, R.W. Farndale, S.M. Best, R.E.

Cameron, Crosslinking and composition influence the surface properties,

mechanical stiffness and cell reactivity of collagen-based films, Acta Biomaterialia

8(8) (2012) 3080-3090.

[234] C.E. Campiglio, N. Contessi Negrini, S. Farè, L. Draghi, Cross-Linking Strategies

for Electrospun Gelatin Scaffolds, Materials (Basel) 12(15) (2019) 2476.

[235] K. Yue, G. Trujillo-de Santiago, M.M. Alvarez, A. Tamayol, N. Annabi, A.

Khademhosseini, Synthesis, properties, and biomedical applications of gelatin

methacryloyl (GelMA) hydrogels, Biomaterials 73 (2015) 254-271.

[236] P.A. Turner, J.S. Thiele, J.P. Stegemann, Growth factor sequestration and enzyme-

mediated release from genipin-crosslinked gelatin microspheres, Journal of

biomaterials science. Polymer edition 28(16) (2017) 1826-1846.

[237] A.H. Nguyen, J. McKinney, T. Miller, T. Bongiorno, T.C. McDevitt, Gelatin

methacrylate microspheres for controlled growth factor release, Acta biomaterialia

13 (2015) 101-110.

[238] P. Jaipan, A. Nguyen, R.J. Narayan, Gelatin-based hydrogels for biomedical

applications, MRS Communications 7(3) (2017) 416-426.

[239] A.P. Kishan, T. Walker, N. Sears, T. Wilems, E.M. Cosgriff‐Hernandez,

Development of a bi‐modal, in situ crosslinking method to achieve multi‐factor

release from electrospun gelatin, Journal of Biomedical Materials Research Part A

(2018).

[240] J. Su, S.C. Satchell, J.A. Wertheim, R.N. Shah, Poly(ethylene glycol)-crosslinked

gelatin hydrogel substrates with conjugated bioactive peptides influence

endothelial cell behavior, Biomaterials 201 (2019) 99-112.

[241] S. Prakash Parthiban, D. Rana, E. Jabbari, N. Benkirane-Jessel, M. Ramalingam,

Covalently immobilized VEGF-mimicking peptide with gelatin methacrylate

enhances microvascularization of endothelial cells, Acta Biomaterialia 51 (2017)

330-340.

[242] A.C. Mitchell, P.S. Briquez, J.A. Hubbell, J.R. Cochran, Engineering growth factors

for regenerative medicine applications, Acta Biomaterialia 30 (2016) 1-12.

Page 200: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

177

[243] Z. Li, T. Qu, C. Ding, C. Ma, H. Sun, S. Li, X. Liu, Injectable gelatin derivative

hydrogels with sustained vascular endothelial growth factor release for induced

angiogenesis, Acta Biomaterialia 13 (2015) 88-100.

[244] C. Claaßen, L. Sewald, G.E.M. Tovar, K. Borchers, Controlled Release of Vascular

Endothelial Growth Factor from Heparin-Functionalized Gelatin Type A and

Albumin Hydrogels, Gels 3(4) (2017) 35.

[245] D.G. Belair, N.N. Le, W.L. Murphy, Design of growth factor sequestering

biomaterials, Chem Commun (Camb) 50(99) (2014) 15651-15668.

[246] S. Pacelli, F. Acosta, A.R. Chakravarti, S.G. Samanta, J. Whitlow, S. Modaresi,

R.P.H. Ahmed, J. Rajasingh, A. Paul, Nanodiamond-based injectable hydrogel for

sustained growth factor release: Preparation, characterization and in vitro analysis,

Acta Biomaterialia 58 (2017) 479-491.

[247] R. Waters, S. Pacelli, R. Maloney, I. Medhi, R.P. Ahmed, A. Paul, Stem cell

secretome-rich nanoclay hydrogel: a dual action therapy for cardiovascular

regeneration, Nanoscale 8(14) (2016) 7371-7376.

[248] J.C. Bragg, H. Kweon, Y. Jo, K.G. Lee, C.-C. Lin, In situ formation of silk-gelatin

hybrid hydrogels for affinity-based growth factor sequestration and release, RSC

Advances 6(115) (2016) 114353-114360.

[249] G. Thakur, D. Rousseau, R.R. Rafanan, Gelatin based matrices for drug delivery

applications, Gelatin: Production, Applications and Health Implications (2013).

[250] H. Wang, O.C. Boerman, K. Sariibrahimoglu, Y. Li, J.A. Jansen, S.C.G.

Leeuwenburgh, Comparison of micro- vs. nanostructured colloidal gelatin gels for

sustained delivery of osteogenic proteins: Bone morphogenetic protein-2 and

alkaline phosphatase, Biomaterials 33(33) (2012) 8695-8703.

[251] J. Lee, J.J. Yoo, A. Atala, S.J. Lee, The effect of controlled release of PDGF-BB

from heparin-conjugated electrospun PCL/gelatin scaffolds on cellular bioactivity

and infiltration, Biomaterials 33(28) (2012) 6709-6720.

[252] M.B. Oliveira, J.F. Mano, Polymer‐based microparticles in tissue engineering and

regenerative medicine, Biotechnology progress 27(4) (2011) 897-912.

[253] K.M.Z. Hossain, U. Patel, I. Ahmed, Development of microspheres for biomedical

applications: a review, Progress in biomaterials 4(1) (2015) 1-19.

[254] M.C. Echave, P. Sánchez, J.L. Pedraz, G. Orive, Progress of gelatin-based 3D

approaches for bone regeneration, Journal of Drug Delivery Science and

Technology 42 (2017) 63-74.

[255] W. Habraken, O. Boerman, J. Wolke, A. Mikos, J. Jansen, In vitro growth factor

release from injectable calcium phosphate cements containing gelatin

microspheres, Journal of Biomedical Materials Research Part A 91(2) (2009) 614-

622.

Page 201: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

178

[256] Y. Li, H. Meng, Y. Liu, A. Narkar, B.P. Lee, Gelatin microgel incorporated

poly(ethylene glycol)-based boadhesive with enhanced adhesive property and

bioactivity, ACS Applied Materials & Interfaces 8(19) (2016) 11980-11989.

[257] V. Gupta, Y. Khan, C.J. Berkland, C.T. Laurencin, M.S. Detamore, Microsphere-

based scaffolds in regenerative engineering, Annual review of biomedical

engineering 19 (2017) 135-161.

[258] X. Zhao, S. Liu, L. Yildirimer, H. Zhao, R. Ding, H. Wang, W. Cui, D. Weitz,

Injectable Stem Cell-Laden Photocrosslinkable Microspheres Fabricated Using

Microfluidics for Rapid Generation of Osteogenic Tissue Constructs, Advanced

Functional Materials 26(17) (2016) 2809-2819.

[259] K.-S. Park, C. Kim, J.-O. Nam, S.-M. Kang, C.-S. Lee, Synthesis and

characterization of thermosensitive gelatin hydrogel microspheres in a microfluidic

system, Macromolecular Research 24(6) (2016) 529-536.

[260] N. Bock, M.A. Woodruff, D.W. Hutmacher, T.R. Dargaville, Electrospraying, a

reproducible method for production of polymeric microspheres for biomedical

applications, Polymers 3(1) (2011) 131-149.

[261] T.A. Holland, Y. Tabata, A.G. Mikos, In vitro release of transforming growth factor-

β1 from gelatin microparticles encapsulated in biodegradable, injectable

oligo(poly(ethylene glycol) fumarate) hydrogels, Journal of Controlled Release

91(3) (2003) 299-313.

[262] Y. Kimura, M. Ozeki, T. Inamoto, Y. Tabata, Adipose tissue engineering based on

human preadipocytes combined with gelatin microspheres containing basic

fibroblast growth factor, Biomaterials 24(14) (2003) 2513-2521.

[263] Y. Luo, D. Li, X. Xie, P. Kang, Porous, lithium-doped calcium polyphosphate

composite scaffolds containing vascular endothelial growth factor (VEGF)-loaded

gelatin microspheres for treating glucocorticoid-induced osteonecrosis of the

femoral head, Biomedical Materials 14(3) (2019) 035013.

[264] K. De Clercq, C. Schelfhout, M. Bracke, O. De Wever, M. Van Bockstal, W. Ceelen,

J.P. Remon, C. Vervaet, Genipin-crosslinked gelatin microspheres as a strategy to

prevent postsurgical peritoneal adhesions: In vitro and in vivo characterization,

Biomaterials 96 (2016) 33-46.

[265] R.T. Annamalai, P.A. Turner, W.F. Carson, B. Levi, S. Kunkel, J.P. Stegemann,

Harnessing macrophage-mediated degradation of gelatin microspheres for

spatiotemporal control of BMP2 release, Biomaterials 161 (2018) 216-227.

[266] H. Zhuang, S. Bu, L. Hua, M.A. Darabi, X. Cao, M. Xing, Gelatin-methacrylamide

gel loaded with microspheres to deliver GDNF in bilayer collagen conduit

promoting sciatic nerve growth, International journal of nanomedicine 11 (2016)

1383.

Page 202: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

179

[267] Y. Zhao, Z. Liu, C. Pan, Z. Li, J. Zhou, J. Wang, Z. Yin, X. Wang, Preparation of

gelatin microspheres encapsulated with bFGF for therapeutic angiogenesis in a

canine ischemic hind limb, Journal of Biomaterials Science, Polymer Edition 22(4-

6) (2011) 665-682.

[268] L.D. Solorio, E.L. Vieregge, C.D. Dhami, P.N. Dang, E. Alsberg, Engineered

cartilage via self-assembled hMSC sheets with incorporated biodegradable gelatin

microspheres releasing transforming growth factor-β1, Journal of Controlled

Release 158(2) (2012) 224-232.

[269] R. Tsaryk, A. Gloria, T. Russo, L. Anspach, R. De Santis, S. Ghanaati, R.E. Unger,

L. Ambrosio, C.J. Kirkpatrick, Collagen-low molecular weight hyaluronic acid

semi-interpenetrating network loaded with gelatin microspheres for cell and growth

factor delivery for nucleus pulposus regeneration, Acta Biomaterialia 20 (2015) 10-

21.

[270] H. Matsumine, R. Sasaki, Y. Tabata, M. Matsui, M. Yamato, T. Okano, H. Sakurai,

Facial nerve regeneration using basic fibroblast growth factor‐impregnated gelatin

microspheres in a rat model, Journal of tissue engineering and regenerative

medicine 10(10) (2016) E559-E567.

[271] T.N. Vo, F.K. Kasper, A.G. Mikos, Strategies for controlled delivery of growth

factors and cells for bone regeneration, Advanced Drug Delivery Reviews 64(12)

(2012) 1292-1309.

[272] I. Banerjee, D. Mishra, T.K. Maiti, PLGA Microspheres Incorporated Gelatin

Scaffold: Microspheres Modulate Scaffold Properties, International Journal of

Biomaterials 2009 (2009) 9.

[273] C.J. Tsao, L. Pandolfi, X. Wang, S. Minardi, C. Lupo, M. Evangelopoulos, T.

Hendrickson, A. Shi, G. Storci, F. Taraballi, Electrospun patch functionalized with

nanoparticles allows for spatiotemporal release of VEGF and PDGF-BB promoting

in vivo neovascularization, ACS applied materials & interfaces 10(51) (2018)

44344-44353.

[274] Y. Zuo, X. He, Y. Yang, D. Wei, J. Sun, M. Zhong, R. Xie, H. Fan, X. Zhang,

Microfluidic-based generation of functional microfibers for biomimetic complex

tissue construction, Acta biomaterialia 38 (2016) 153-162.

[275] J. Rickman, G. Tronci, H. Liang, S.J. Russell, Rotation-assisted wet-spinning of UV-

cured gelatin fibres and nonwovens, Journal of Materials Science 54(14) (2019)

10529-10547.

[276] G. An, W. Zhang, D. Ma, B. Lu, G. Wei, Y. Guang, C. Ru, Y. Wang, Influence of

VEGF/BMP-2 on the proliferation and osteogenetic differentiation of rat bone

mesenchymal stem cells on PLGA/gelatin composite scaffold, Eur Rev Med

Pharmacol Sci 21(10) (2017) 2316-2328.

Page 203: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

180

[277] M. Norouzi, I. Shabani, H.H. Ahvaz, M. Soleimani, PLGA/gelatin hybrid

nanofibrous scaffolds encapsulating EGF for skin regeneration, Journal of

biomedical materials research Part A 103(7) (2015) 2225-2235.

[278] D.C. Ardila, E. Tamimi, T. Doetschman, W.R. Wagner, J.P. Vande Geest,

Modulating smooth muscle cell response by the release of TGFβ2 from tubular

scaffolds for vascular tissue engineering, Journal of Controlled Release 299 (2019)

44-52.

[279] J. Wu, Y. Hong, Enhancing cell infiltration of electrospun fibrous scaffolds in tissue

regeneration, Bioact Mater 1(1) (2016) 56-64.

[280] Z. Tan, X. Gao, T. Liu, Y. Yang, J. Zhong, C. Tong, Y. Tan, Electrospun vein grafts

with high cell infiltration for vascular tissue engineering, Materials Science and

Engineering: C 81 (2017) 407-415.

[281] A.A. Ucuzian, A.A. Gassman, A.T. East, H.P. Greisler, Molecular mediators of

angiogenesis, Journal of Burn Care & Research 31(1) (2010) 158-175.

[282] Y.-H. Yu, D. Lee, Y.-H. Hsu, Y.-C. Chou, S.W. Ueng, C.-K. Chen, S.-J. Liu, A

Three-Dimensional Printed Polycaprolactone Scaffold Combined with Co-Axially

Electrospun Vancomycin/Ceftazidime/Bone Morphological Protein-2 Sheath-Core

Nanofibers for the Repair of Segmental Bone Defects During the Masquelet

Procedure, International journal of nanomedicine 15 (2020) 913-925.

[283] Y.-H. Yu, R.-C. Wu, D. Lee, C.-K. Chen, S.-J. Liu, Artificial membrane induced by

novel biodegradable nanofibers in the Masquelet procedure for treatment of

segmental bone defects, Journal of Nanomaterials 2018 (2018).

[284] M. Shin, H. Yoshimoto, J.P. Vacanti, In vivo bone tissue engineering using

mesenchymal stem cells on a novel electrospun nanofibrous scaffold, Tissue

engineering 10(1-2) (2004) 33-41.

[285] A.K. Ekaputra, Y. Zhou, S.M. Cool, D.W. Hutmacher, Composite electrospun

scaffolds for engineering tubular bone grafts, Tissue Engineering Part A 15(12)

(2009) 3779-3788.

[286] Z. Guo, S. Wu, H. Li, Q. Li, G. Wu, C. Zhou, In vitro evaluation of electrospun

PLGA/PLLA/PDLLA blend fibers loaded with naringin for guided bone

regeneration, Dental materials journal (2018) 2016-220.

[287] S.H. Lim, H.-Q. Mao, Electrospun scaffolds for stem cell engineering, Advanced

Drug Delivery Reviews 61(12) (2009) 1084-1096.

[288] H. Zigdon-Giladi, A. Khutaba, R. Elimelech, E.E. Machtei, S. Srouji, VEGF release

from a polymeric nanofiber scaffold for improved angiogenesis, Journal of

biomedical materials research. Part A 105(10) (2017) 2712-2721.

Page 204: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

181

[289] C. Del Gaudio, S. Baiguera, M. Boieri, B. Mazzanti, D. Ribatti, A. Bianco, P.

Macchiarini, Induction of angiogenesis using VEGF releasing genipin-crosslinked

electrospun gelatin mats, Biomaterials 34(31) (2013) 7754-65.

[290] A. Kishan, T. Walker, N. Sears, T. Wilems, E. Cosgriff‐Hernandez, Development of

a bimodal, in situ crosslinking method to achieve multifactor release from

electrospun gelatin, Journal of Biomedical Materials Research Part A 106(5) (2018)

1155-1164.

[291] A. Ovsianikov, A. Deiwick, S. Van Vlierberghe, P. Dubruel, L. Moller, G. Drager,

B. Chichkov, Laser fabrication of three-dimensional CAD scaffolds from

photosensitive gelatin for applications in tissue engineering, Biomacromolecules

12(4) (2011) 851-858.

[292] T. Billiet, B.V. Gasse, E. Gevaert, M. Cornelissen, J.C. Martins, P. Dubruel,

Quantitative Contrasts in the Photopolymerization of Acrylamide and

Methacrylamide‐Functionalized Gelatin Hydrogel Building Blocks,

Macromolecular bioscience 13(11) (2013) 1531-1545.

[293] E. Groppa, S. Brkic, E. Bovo, S. Reginato, V. Sacchi, N. Di Maggio, M.G. Muraro,

D. Calabrese, M. Heberer, R. Gianni‐Barrera, VEGF dose regulates vascular

stabilization through Semaphorin3A and the Neuropilin‐1+ monocyte/TGF‐β1

paracrine axis, EMBO molecular medicine 7(10) (2015) 1366-1384.

[294] K. Xu, O. Cleaver, Tubulogenesis during blood vessel formation, Semin Cell Dev

Biol 22(9) (2011) 993-1004.

[295] G. Carpentier, M. Martinelli, J. Courty, I. Cascone, Angiogenesis analyzer for

ImageJ, 4th ImageJ User and Developer Conference proceedings, 2012, pp. 198-

201.

[296] M.D. Hoffman, D.S. Benoit, Emulating native periosteum cell population and

subsequent paracrine factor production to promote tissue engineered periosteum-

mediated allograft healing, Biomaterials 52 (2015) 426-440.

[297] S.R. Shah, B.T. Smith, A.M. Tatara, E.R. Molina, E.J. Lee, T.C. Piepergerdes, B.A.

Uhrig, R.E. Guldberg, G.N. Bennett, J.C. Wenke, A.G. Mikos, Effects of Local

Antibiotic Delivery from Porous Space Maintainers on Infection Clearance and

Induction of an Osteogenic Membrane in an Infected Bone Defect, Tissue

Engineering Part A 23(3-4) (2016) 91-100.

[298] N. Davidenko, C.F. Schuster, D.V. Bax, R.W. Farndale, S. Hamaia, S.M. Best, R.E.

Cameron, Evaluation of cell binding to collagen and gelatin: a study of theeffect of

2D and 3D architecture and surface chemistry, Journal of Materials Science:

Materials in Medicine 27(10) (2016) 148.

[299] C. Del Gaudio, S. Baiguera, M. Boieri, B. Mazzanti, D. Ribatti, A. Bianco, P.

Macchiarini, Induction of angiogenesis using VEGF releasing genipin-crosslinked

electrospun gelatin mats, Biomaterials 34(31) (2013) 7754-7765.

Page 205: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

182

[300] M. Rajabi, S.A. Mousa, The role of angiogenesis in cancer treatment, Biomedicines

5(2) (2017) 34.

[301] L. Wu, Y. Gu, L. Liu, J. Tang, J. Mao, K. Xi, Z. Jiang, Y. Zhou, Y. Xu, L. Deng, L.

Chen, W. Cui, Hierarchical micro/nanofibrous membranes of sustained releasing

VEGF for periosteal regeneration, Biomaterials 227 (2020) 119555.

[302] H. Zhang, X. Jia, F. Han, J. Zhao, Y. Zhao, Y. Fan, X. Yuan, Dual-delivery of VEGF

and PDGF by double-layered electrospun membranes for blood vessel

regeneration, Biomaterials 34(9) (2013) 2202-2212.

[303] S.A. Theron, A.L. Yarin, E. Zussman, E. Kroll, Multiple jets in electrospinning:

experiment and modeling, Polymer 46(9) (2005) 2889-2899.

[304] H. Lee, S.M. Dellatore, W.M. Miller, P.B. Messersmith, Mussel-inspired surface

chemistry for multifunctional coatings, science 318(5849) (2007) 426-430.

[305] Y.H. Ding, M. Floren, W. Tan, Mussel-inspired polydopamine for bio-surface

functionalization, Biosurface and Biotribology 2(4) (2016) 121-136.

[306] S.H. Ku, C.B. Park, Human endothelial cell growth on mussel-inspired nanofiber

scaffold for vascular tissue engineering, Biomaterials 31(36) (2010) 9431-9437.

[307] H. Lee, S.M. Dellatore, W.M. Miller, P.B. Messersmith, Mussel-Inspired Surface

Chemistry for Multifunctional Coatings, Science 318(5849) (2007) 426.

[308] S. Shiels, D. Tennent, A. Lofgren, J. Wenke, Topical rifampin powder for

orthopaedic trauma part II: Topical rifampin allows for spontaneous bone healing

in sterile and contaminated wounds: TOPICAL RIFAMPIN ALLOWS

FRACTURE HEALING, Journal of Orthopaedic Research 36 (2018).

[309] M.D. Hoffman, D.S.W. Benoit, Emerging ideas: Engineering the periosteum:

revitalizing allografts by mimicking autograft healing, Clin Orthop Relat Res

471(3) (2013) 721-726.

[310] M.D. Hoffman, D.S.W. Benoit, Emulating native periosteum cell population and

subsequent paracrine factor production to promote tissue engineered periosteum-

mediated allograft healing, Biomaterials 52 (2015) 426-440.

[311] M.P. Mingeot-Leclercq, P.M. Tulkens, Aminoglycosides: nephrotoxicity,

Antimicrobial agents and chemotherapy 43(5) (1999) 1003-1012.

[312] P. Gentile, V. Chiono, I. Carmagnola, P.V. Hatton, An overview of poly(lactic-co-

glycolic) acid (PLGA)-based biomaterials for bone tissue engineering, Int J Mol

Sci 15(3) (2014) 3640-3659.

[313] C.f.D. Control, Prevention, CDC/NCHS National Hospital Discharge Survey, 2010.

Number and rate of discharges from short-stay hospitals and days of care, with

average length of stay and standard error, by selected first-listed diagnostic

categories: United States, 2010. CDC Web site, 2013.

Page 206: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

183

[314] B. Davis, D.E. Rivadeneira, Complications of colorectal anastomoses, Surgical

Clinics 93(1) (2013) 61-87.

[315] J. Hammond, S. Lim, Y. Wan, X. Gao, A. Patkar, The burden of gastrointestinal

anastomotic leaks: an evaluation of clinical and economic outcomes, Journal of

Gastrointestinal Surgery 18(6) (2014) 1176-1185.

[316] N. Hyman, T.L. Manchester, T. Osler, B. Burns, P.A. Cataldo, Anastomotic leaks

after intestinal anastomosis: it's later than you think, Annals of surgery 245(2)

(2007) 254.

[317] J.W. Ogilvie, D.W. Dietz, L. Stocchi, Anastomotic leak after restorative

proctosigmoidectomy for cancer: what are the chances of a permanent ostomy?,

International journal of colorectal disease 27(10) (2012) 1259-1266.

[318] B.C. Paun, S. Cassie, A.R. MacLean, E. Dixon, W.D. Buie, Postoperative

complications following surgery for rectal cancer, Annals of surgery 251(5) (2010)

807-818.

[319] J.M. Daly, J.J. DeCosse, Complications in surgery of the colon and rectum, Surgical

Clinics of North America 63(6) (1983) 1215-1231.

[320] N. Bölgen, I. Vargel, P. Korkusuz, Y.Z. Menceloğlu, E. Pişkin, In vivo performance

of antibiotic embedded electrospun PCL membranes for prevention of abdominal

adhesions, Journal of Biomedical Materials Research Part B: Applied Biomaterials

81(2) (2007) 530-543.

[321] A.H. DeCherney, Cllinical problem of intraperitoneal postsurgical adhesion

formation following generaly surgery and the use of adheson prevention barriers

Surgical Clinics of North America 77(3) (1997) 671-688.

[322] T.Z. Nursal, R. Anarat, S. Bircan, S. Yildirim, A. Tarım, M. Haberal, The effect of

tissue adhesive, octyl-cyanoacrylate, on the healing of experimental high-risk and

normal colonic anastomoses, The American journal of surgery 187(1) (2004) 28-

32.

[323] A.C. van der Ham, W.J. Kort, I.M. Weijma, H. Jeekel, Transient protection of

incomplete colonic anastomoses with fibrin sealant: an experimental study in the

rat, Journal of Surgical Research 55(3) (1993) 256-260.

[324] A. Van der Ham, W. Kort, I. Weijma, V. Ingh, H. Den, J. Jeekel, Effect of fibrin

sealant on the healing colonic anastomosis in the rat, British journal of surgery

78(1) (1991) 49-53.

[325] P. Senthil‐Kumar, T. Ni, M.A. Randolph, G.C. Velmahos, I.E. Kochevar, R.W.

Redmond, A light‐activated amnion wrap strengthens colonic anastomosis and

reduces peri‐anastomotic adhesions, Lasers in surgery and medicine 48(5) (2016)

530-537.

Page 207: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

184

[326] A. Agnifili, M. Schietroma, A. Carloni, S. Mattucci, G. Caterino, N. Lygidakis, F.

Carlei, The value of omentoplasty in protecting colorectal anastomosis from

leakage. A prospective randomized study in 126 patients, Hepato-gastroenterology

51(60) (2004) 1694-1697.

[327] F. Merad, J.-M. Hay, A. Fingerhut, Y. Flamant, J.-M. Molkhou, Y. Laborde,

Omentoplasty in the prevention of anastomotic leakage after colonic or rectal

resection: a prospective randomized study in 712 patients. French Associations for

Surgical Research, Annals of surgery 227(2) (1998) 179.

[328] G.F. Hagerman, W.B. Gaertner, G.R. Ruth, M.L. Potter, R.E. Karulf, Bovine

pericardium buttress reinforces colorectal anastomoses in a canine model, Diseases

of the colon & rectum 50(7) (2007) 1053-1060.

[329] J. Hoeppner, K. Willa, S. Timme, D. Tittelbach-Helmrich, U. Hopt, T. Keck, G.

Marjanovic, Reinforcement of colonic anastomoses with a collagenous double-

layer matrix extracted from porcine dermis, European Surgical Research 45(2)

(2010) 68-76.

[330] M. Uludag, B. Citgez, O. Ozkaya, G. Yetkin, O. Ozcan, N. Polat, A. Isgor, Effects

of amniotic membrane on the healing of normal and high-risk colonic anastomoses

in rats, International journal of colorectal disease 24(7) (2009) 809.

[331] J. Hoeppner, V. Crnogorac, G. Marjanovic, E. Jüttner, T. Keck, H.-F. Weiser, U.T.

Hopt, Small intestinal submucosa for reinforcement of colonic anastomosis,

International journal of colorectal disease 24(5) (2009) 543-550.

[332] J. Hoeppner, B. Wassmuth, G. Marjanovic, S. Timme, U.T. Hopt, T. Keck,

Anastomotic sealing by extracellular matrices (ECM) improves healing of colonic

anastomoses in the critical early phase, Journal of Gastrointestinal Surgery 14(6)

(2010) 977-986.

[333] S. Matsuda, N. Se, H. Iwata, Y. Ikada, Evaluation of the antiadhesion potential of

UV cross-linked gelatin films in a rat abdominal model, Biomaterials 23(14) (2002)

2901-2908.

[334] J.M. Becker, M.T. Dayton, V.W. Fazio, D.E. Beck, S.J. Stryker, S.D. Wexner, B.G.

Wolff, P.L. Roberts, L.E. Smith, S.A. Sweeney, Prevention of postoperative

abdominal adhesions by a sodium hyaluronate-based bioresorbable membrane: a

prospective, randomized, double-blind multicenter study, Journal of the American

College of surgeons 183(4) (1996) 297-306.

[335] A.B.S. Group, Prevention of postsurgical adhesions by INTERCEED (TC7), an

absorbable adhesion barrier: a prospective randomized multicenter clinical study,

Fertil Steril 51(6) (1989) 933-8.

[336] A. Hirschelmann, G. Tchartchian, M. Wallwiener, A. Hackethal, R.L. De Wilde, A

review of the problematic adhesion prophylaxis in gynaecological surgery, Arch

Gynecol Obstet 285(4) (2012) 1089-1097.

Page 208: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

185

[337] D. Wiseman, L. Gottlick-Iarkowski, L. Kamp, Effect of different barriers of oxidized

regenerated cellulose (ORC) on cecal and sidewall adhesions in the presence and

absence of bleeding, Journal of Investigative Surgery 12(3) (1999) 141-146.

[338] Q. Zeng, Z. Yu, J. You, Q. Zhang, Efficacy and safety of Seprafilm for preventing

postoperative abdominal adhesion: systematic review and meta-analysis, World

journal of surgery 31(11) (2007) 2125-2131.

[339] D.E. Beck, Z. Cohen, J.W. Fleshman, H.S. Kaufman, H. van Goor, B.G. Wolff, A

prospective, randomized, multicenter, controlled study of the safety of Seprafilm®

adhesion barrier in abdominopelvic surgery of the intestine, Diseases of the colon

& rectum 46(10) (2003) 1310-1319.

[340] T. Horii, H. Tsujimoto, H. Miyamoto, K. Yamanaka, S. Tanaka, H. Torii, Y.

Ozamoto, H. Takamori, E. Nakamachi, Y. Ikada, Physical and biological properties

of a novel anti‐adhesion material made of thermally cross‐linked gelatin film:

Investigation of the usefulness as anti‐adhesion material, Journal of Biomedical

Materials Research Part B: Applied Biomaterials (2017).

[341] A. Kishan, R. Nezarati, C. Radzicki, A. Renfro, J. Robinson, M. Whitely, E. Cosgriff-

Hernandez, In situ crosslinking of electrospun gelatin for improved fiber

morphology retention and tunable degradation, Journal of Materials Chemistry B

3(40) (2015) 7930-7938.

[342] S. Cereceres, T. Touchet, M.B. Browning, C. Smith, J. Rivera, M. Höök, C.

Whitfield-Cargile, B. Russell, E. Cosgriff-Hernandez, Chronic wound dressings

based on collagen-mimetic proteins, Adv Wound Care (New Rochelle) 4(8) (2015)

444-456.

[343] M.S. Hahn, M.K. McHale, E. Wang, R.H. Schmedlen, J.L. West, Physiologic

Pulsatile Flow Bioreactor Conditioning of Poly(ethylene glycol)-based Tissue

Engineered Vascular Grafts, Annals of Biomedical Engineering 35(2) (2007) 190-

200.

[344] M.B. Browning, E. Cosgriff-Hernandez, Development of a biostable replacement for

PEGDA hydrogels, Biomacromolecules 13(3) (2012) 779-786.

[345] S. Cereceres, Z. Lan, L. Bryan, M. Whitely, T. Wilems, N. Fabela, C. Whitfield, E.

Cosgriff-Hernandez, In Vivo Characterization of Poly(ethylene glycol) Hydrogels

with Thio-β Esters, Annals of Biomedical Engineering 48 (2019).

[346] M. Deshmukh, Y. Singh, S. Gunaseelan, D. Gao, S. Stein, P.J. Sinko, Biodegradable

poly(ethylene glycol) hydrogels based on a self-elimination degradation

mechanism, Biomaterials 31(26) (2010) 6675-6684.

[347] S.-C. Wu, W.-H. Chang, G.-C. Dong, K.-Y. Chen, Y.-S. Chen, C.-H. Yao, Cell

adhesion and proliferation enhancement by gelatin nanofiber scaffolds, Journal of

Bioactive and Compatible Polymers 26(6) (2011) 565-577.

Page 209: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

186

[348] G.B. Schneider, A. English, M. Abraham, R. Zaharias, C. Stanford, J. Keller, The

effect of hydrogel charge density on cell attachment, Biomaterials 25(15) (2004)

3023-3028.

[349] M. Wancura, M. Talanker, S. Toubbeh, A. Bryan, E. Cosgriff-Hernandez, Bioactive

hydrogel coatings of complex substrates using diffusion-mediated redox initiation,

Journal of Materials Chemistry B 8(19) (2020) 4289-4298.

[350] D. Poehnert, M. Abbas, H.H. Kreipe, J. Klempnauer, M. Winny, High reproducibility

of adhesion formation in rat with meso-stitch approximation of injured cecum and

abdominal wall, International journal of medical sciences 12(1) (2015) 1-6.

[351] J. Jiang, W. Wan, L. Ge, S. Bu, W. Zhong, M. Xing, Mussel-inspired nanofibrous

sheet for suture-less stomach incision surgery, Chemical Communications 51(41)

(2015) 8695-8698.

[352] Y. Liu, M.B. Chan-Park, Hydrogel based on interpenetrating polymer networks of

dextran and gelatin for vascular tissue engineering, Biomaterials 30(2) (2009) 196-

207.

[353] A.K. Davey, P.J. Maher, Surgical adhesions: a timely update, a great challenge for

the future, Journal of minimally invasive gynecology 14(1) (2007) 15-22.

[354] D. Ntourakis, M. Katsimpoulas, A. Tanoglidi, C. Barbatis, P.E. Karayannacos, T.N.

Sergentanis, N. Kostomitsopoulos, A. Machairas, Adhesions and Healing of

Intestinal Anastomoses:The Effect of Anti-Adhesion Barriers, Surgical Innovation

23(3) (2016) 266-276.

[355] D.-J. Yang, F. Chen, Z.-C. Xiong, C.-D. Xiong, Y.-Z. Wang, Tissue anti-adhesion

potential of biodegradable PELA electrospun membranes, Acta biomaterialia 5(7)

(2009) 2467-2474.

[356] X.M. Mo, C.Y. Xu, M. Kotaki, S. Ramakrishna, Electrospun P(LLA-CL) nanofiber:

a biomimetic extracellular matrix for smooth muscle cell and endothelial cell

proliferation, Biomaterials 25(10) (2004) 1883-1890.

[357] Y.S. Choi, S.R. Hong, Y.M. Lee, K.W. Song, M.H. Park, Y.S. Nam, Studies on

gelatin-containing artificial skin: II. Preparation and characterization of cross-

linked gelatin-hyaluronate sponge, Journal of Biomedical Materials Research 48(5)

(1999) 631-639.

[358] J.L. West, J.A. Hubbell, Comparison of covalently and physically cross-linked

polyethylene glycol-based hydrogels for the prevention of postoperative adhesions

in a rat model, Biomaterials 16(15) (1995) 1153-1156.

[359] M.B. Browning, S.N. Cereceres, P.T. Luong, E.M. Cosgriff-Hernandez,

Determination of the in vivo degradation mechanism of PEGDA hydrogels, Journal

of biomedical materials research. Part A 102(12) (2014) 4244-4251.

Page 210: Copyright by Taneidra Walker Buie 2020 - Front Matter Template

187

[360] S. Cereceres, Z. Lan, L. Bryan, M. Whitely, T. Wilems, N. Fabela, C. Whitfield-

Cargile, E. Cosgriff-Hernandez, In Vivo Characterization of Poly(ethylene glycol)

Hydrogels with Thio-β Esters, Annals of Biomedical Engineering 48(3) (2019)

953-967.

[361] S. Kovačič, E. Žagar, C. Slugovc, Strength versus toughness of emulsion templated

Poly(Dicyclopentadiene) foams, Polymer 169 (2019) 58-65.

[362] Y. Wan, Q. Wu, S. Wang, S. Zhang, Z. Hu, Mechanical Properties of Porous

Polylactide/Chitosan Blend Membranes, Macromolecular Materials and

Engineering 292(5) (2007) 598-607.

[363] M. Browning, T. Wilems, M. Hahn, E. Cosgriff‐Hernandez, Compositional control

of poly (ethylene glycol) hydrogel modulus independent of mesh size, Journal of

Biomedical Materials Research Part A 98(2) (2011) 268-273.

[364] C.B. Hutson, J.W. Nichol, H. Aubin, H. Bae, S. Yamanlar, S. Al-Haque, S.T. Koshy,

A. Khademhosseini, Synthesis and characterization of tunable poly (ethylene

glycol): gelatin methacrylate composite hydrogels, Tissue Engineering Part A

17(13-14) (2011) 1713-1723.

[365] S.H. Bae, S.R. Son, S. Kumar Sakar, T.H. Nguyen, S.W. Kim, Y.K. Min, B.T. Lee,

Evaluation of the potential anti‐adhesion effect of the PVA/Gelatin membrane,

Journal of Biomedical Materials Research Part B: Applied Biomaterials 102(4)

(2014) 840-849.

[366] Y. Li, H. Meng, Y. Liu, A. Narkar, B.P. Lee, Gelatin Microgel Incorporated

Poly(ethylene glycol)-Based Bioadhesive with Enhanced Adhesive Property and

Bioactivity, ACS applied materials & interfaces 8(19) (2016) 11980-11989.

[367] J. Zhang, A. Mujeeb, J. Feng, Y. Li, Y. Du, J. Lin, Z. Ge, Physically entrapped gelatin

in polyethylene glycol scaffolds for three-dimensional chondrocyte culture, Journal

of Bioactive and Compatible Polymers 31(5) (2016) 513-530.

[368] M. Mehdizadeh, H. Weng, D. Gyawali, L. Tang, J. Yang, Injectable citrate-based

mussel-inspired tissue bioadhesives with high wet strength for sutureless wound

closure, Biomaterials 33(32) (2012) 7972-7983.

[369] H. Zhang, L.P. Bré, T. Zhao, Y. Zheng, B. Newland, W. Wang, Mussel-inspired

hyperbranched poly (amino ester) polymer as strong wet tissue adhesive,

Biomaterials 35(2) (2014) 711-719.

[370] M. Mehdizadeh, J. Yang, Design strategies and applications of tissue bioadhesives,

Macromolecular bioscience 13(3) (2013) 271-288.