411
Determining the Oncological and Immunological Effects of Histotripsy for Tumor Ablation Alissa D Hendricks-Wenger Dissertation submitted to the faculty of the Virginia Polytechnic Institute and State University in partial fulfillment of the requirements for the degree of Doctor of Philosophy In Translational Biology, Medicine and Health Eli Vlaisavljevich Irving Coy Allen Eva Schmelz Scott Verbridge David Luyimbazi April 26, 2021 Blacksburg, VA Keywords: histotripsy, ablation, cancer, immunology, oncology models This work is licensed under a Creative Commons Attribution 4.0 International License by Alissa Danielle Hendricks-Wenger under http://creativecommons.org/licenses/by/4.0

Determining the Oncological and Immunological Effects of

  • Upload
    others

  • View
    5

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Determining the Oncological and Immunological Effects of

Determining the Oncological and Immunological Effects of Histotripsy for Tumor Ablation

Alissa D Hendricks-Wenger

Dissertation submitted to the faculty of the Virginia Polytechnic Institute and State University in partial fulfillment of the requirements for the degree of

Doctor of Philosophy In

Translational Biology, Medicine and Health

Eli Vlaisavljevich Irving Coy Allen

Eva Schmelz Scott Verbridge David Luyimbazi

April 26, 2021 Blacksburg, VA

Keywords: histotripsy, ablation, cancer, immunology, oncology models

This work is licensed under a Creative Commons Attribution 4.0 International

License by Alissa Danielle Hendricks-Wenger under http://creativecommons.org/licenses/by/4.0

Page 2: Determining the Oncological and Immunological Effects of

DETERMINING THE ONCOLOGICAL AND IMMUNOLOGICAL EFFECTS OF

HISTOTRIPSY FOR TUMOR ABLATION

Alissa D Hendricks-Wenger

Abstract

Histotripsy is an emerging non-invasive, non-thermal, image-guided cancer

ablation modality that has recently been approved for its first clinical trial in the United

States (NCT04573881). Histotripsy utilizes focused ultrasound to generate acoustic

cavitation within a tumor to mechanically fractionate targeted tissues. While pre-clinical

work has demonstrated the feasibility of applying histotripsy to solid tumors including

primary liver and renal tumors, there is still a need to investigate the potential of

histotripsy to treat additional malignancies. In investigating the potential for treating

other malignancies there are two avenues that need to be considered: 1) the feasibility

for treating tissues with more complex stromal structures and 2) the ability of histotripsy

to modulate the tumor microenvironment. To determine the safety and feasibility of

additional applications of histotripsy, we conducted dose studies ex vivo on human

tumors and human liver to establish dosimetry metrics for applying histotripsy to more

fibrotic tumors such as cholangiocarcinoma while sparing nearby critical structures,

such as bile ducts and blood vessels. Learning the safety dose-margins from the

excised tissues, we performed an in vivo study using mice bearing patient-derived

xenograft cholangiocarcinoma tumors. With this model, we were able to demonstrate

our ability ablate the stiff cholangiocarcinoma tumors without causing any debilitating

Page 3: Determining the Oncological and Immunological Effects of

off- target damage. To gain a more robust understanding of the effects of histotripsy

ablation on potentially difficult to treat tumors, we developed a porcine xenograft tumor

model and utilized veterinary cancer patients. These studies have helped established

protocols for utilizing histotripsy with ultrasound guidance to treat tumors that are more

difficult to treat and can withstand mechanical ablation, including pancreatic

adenocarcinoma, osteosarcomas, and soft tissue sarcomas. Pigs share many

similarities with human anatomy and physiology, making them an ideal model organism

for testing new medical devices and regimes for treating new targets. Using pigs, we

were able to establish a procedure to utilize histotripsy to target the pancreas in vivo

without causing any lasting or major side effects, such as off-target damage or

pancreatitis. One limitation to the porcine model and veterinary patients, is the limitation

of gaining rapid insight into the immunological effects of histotripsy. Established cancer

mouse models offer the opportunity to rapidly test many organisms with an intact

immune system. We used these mice to study pancreatic adenocarcinoma to determine

the immune response after histotripsy ablation. For these tumors the general response

was an increase in immune cell infiltration post-treatment and a shift in the tumor

microenvironment to a more anti-tumor environment. The results of this dissertation

provide insight into establishing protocols for treating new types of tumors with

histotripsy and immunological effects that lay groundwork for improving future co-

therapeutic treatment planning. Future work will aim to translate histotripsy into clinical

applications and determining co-therapies that can help control metastasis.

Page 4: Determining the Oncological and Immunological Effects of

DETERMINING THE ONCOLOGICAL AND IMMUNOLOGICAL EFFECTS OF

HISTOTRIPSY FOR TUMOR ABLATION

Alissa D Hendricks-Wenger

General Audience Abstract

Histotripsy is a new medical therapy that can remove tumors without the need for

surgery, with the first clinical trial in the United States starting this year, 2021. This

therapy uses focused ultrasound waves to generate powerful microscopic bubbles that

can rapidly destroy targeted tissues with a high-degree of precision. Early studies on

histotripsy have demonstrated the ability of histotripsy to ablate tumors of the liver and

kidneys. In order to be able to fully use this therapy on more difficult to target and treat

cancers more studies are needed. Given that histotripsy uses physical forces to destroy

targets, stronger, more fibrotic tumors and cancers that have begun to spread

throughout the body will be more difficult to treat will need more than simple tumor

removal to better treat these patients. Therefore, when investigating new cancer

applications of histotripsy, it is important to consider the physical features of the tumors

as well as the ability of histotripsy to initiate an immune response against the cancer. To

determine the safety and feasibility of additional applications of histotripsy, we

conducted dose studies on excised human tumors and human liver to see what doses

of histotripsy are required to ablate stronger tumors, such as bile duct tumors. Learning

the potential safety margins of doses from the excised tissues, we conducted a study

using a mouse model to grow stiff, human tumors. With this model, we were able to

Page 5: Determining the Oncological and Immunological Effects of

show that it is possible to ablate the stiffer tumors without causing any major off-target

damage. While it is useful to prove in excised tissues and mice that we can treat certain

tumors, there is an additional need to study the therapy in a model that is more similar

in size and anatomy to humans. Therefore, to gain a better understanding of the effects

of histotripsy on potentially difficult to target and ablate tumors, we developed a novel

porcine tumor model that can support the growth of human tumors and utilized

veterinary cancer patients. These studies have helped established protocols for utilizing

histotripsy to treat difficult to physically ablate tumors and difficult to ultrasound target

tumors, including pancreatic and bone cancers. Established cancer mouse models offer

the opportunity to rapidly test many organisms with an intact immune system. We used

these mice to study pancreatic cancer to determine the immune response after

histotripsy ablation. For this tumor type, while there were slight differences, the general

response was an increase in immune cell infiltration of the tumors post-treatment and a

shift to a stronger immune response against the tumor. The results of this dissertation

provide insight into establishing protocols for treating new types of tumors with

histotripsy and immune effects that lay groundwork for improving future co-therapeutic

planning. Future work will aim to translate histotripsy into clinical applications and

determining co-therapies that can help control body-wide disease.

Page 6: Determining the Oncological and Immunological Effects of

vii

Dedication

To every person and dog that put up with my workaholic

personality, which allowed this body of work happen.

Thanks y’all.

Page 7: Determining the Oncological and Immunological Effects of

viii

Acknowledgements

First and foremost, I would like to thank the first two who set me on the path that

is my PhD research. My thesis advisors Dr. Coy Allen and Dr. Eli Vlaisavljevich. Thanks

to Drs. Allen and Eli I have been able to accomplish a lot and grow as an independent

scientist. Unbeknownst to them (until reading this), I always joked that the only thing

that these two had in common was the PhDs at the end of their names, and this was my

saving grace for the past three years. While this caused me stress from time-to-time,

whenever I was stressed or frustrated, one of them always said what I needed (or

wanted) to hear.

I would also like to thank my current committee Dr. Eva Schmelz, Dr. David

Luyimbazi, and Dr. Scott Verbridge as well as Dr. Kenneth Oestrich, who was a

member of my committee until he moved to Ohio. I appreciate all the time that my

current and past committee members have set aside to listen to my research progress,

offer constructive critiques, and pose thought provoking questions.

Of course, I need to acknowledge the help and support that I have revied over

the years from the other students from both of my labs. I cannot image having survived

the past few years without everyone. It has really been like having two families to get

through the highs and lows of graduate school and real life. From the

engineering/TUSL/Eli/Vlaisavljevich lab (or whatever its being called now), thanks to

everyone who has helped with my projects or just been a person to chat with! Special

thanks to Alex Simon, who started working with histotripsy when I did, and whom I am

100% positive that without his help in the lab and out I would not have been able to get

Page 8: Determining the Oncological and Immunological Effects of

ix

even half of this work done nor would it have been done with half the sanity. Thanks to

Lauren Arnold and Jessica Gannon who are obviously great engineers and both

inspirational, but also were never too busy to help improve all our days by complaining

about how busy we all were. I can also never forget to give a special shout out to Ruby

Hutchinson. While we never really worked together until she was walking out the door,

our ‘hikes’/glorified outdoor pandemic-safe walks and long conversations are

irreplaceable. From the Allen mucosal and cancer immunology lab, thanks to everyone

for helping with work as well as making work-life-balance a normal part of life. Special

thanks to Dr. Rebecca Brock, who started the TBMH program when I did and was an

impeccable roommate for 3 years. By joining the Allen lab first, Becca was the reason

that I met with Dr. Allen when looking to change labs and was therefore able to find my

graduate school niche. Additional thanks to Holly Morrison and Margaret Nagai-Singer

for always finding the positive outlook to difficult situations and being open to presenting

at yet another conference. Finally, of course, thanks to all the undergraduate and

medical students within both groups who have really been a crucial part of why I have

been able to complete this body of work. This especially goes out to Jackie Sereno,

Allison Zeher, Sofie Saunier, Jessica Gannon, Ruby Hutchison, and Pete Weber.

In addition to everyone within my labs, I also need to thank all our collaborators.

Thanks to the veterinary team, including Dr. Sheryl-Coutermarsh Ott, Dr. Joanne Touhy,

Dr. Nick Dervisis, and Dr. Shawna Klahn. Thanks to the various biomedical engineering

teams including Dr. Vincent Wang and his PhD student Chitra Meduri who have helped

with tissue mechanics work and Dr. Rafael Davalos and his PhD students Dr. Natalie

Beitel-White, Dr. Melvin Lorenzo, and Kenneth Aycock who assisted with the IRE

Page 9: Determining the Oncological and Immunological Effects of

x

applications. Thanks to the clinicians from Carillion Clinic Dr. Douglas Grider and Dr.

David Luyimbazi, at Michigan Medicine Dr. Mishal Mendiratta-Lala, and at the

Universitat de Barcelona Dr. Joan Vidal-Jové.

I would also like to thank all of TBMH. Without Jay Read and Becky Langford I

would have been lost in the sea of paperwork. Also, thanks to Dr. Michael Friedlander,

Dr. Audra VanWort, Dr. Steve Poelzing, and Dr. Michelle Theus the directors of TBMH

during my time in the program. Additionally, I would like to thank all the students from

my cohort who helped me during our first-year courses and while preparing for our

qualifying exams.

Of course, an obligatory thanks to my husband Will and our dog Bunny, who put

up with some phases of neglect when I became a bit of a work-o-holic and still love me.

In general, thanks to all my friends and family who have been supportive and loving

throughout this whole process.

Page 10: Determining the Oncological and Immunological Effects of

xi

Table of Contents

Abstract .......................................................................................................................... ii

General Audience Abstract ......................................................................................... iv

Dedication .................................................................................................................... vii

Acknowledgements .................................................................................................... viii

Table of Contents ......................................................................................................... xi

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

List of Tables ........................................................................................................... xxxii

List of Abbreviations .............................................................................................. xxxiii

Attributions for Co-Authored Papers ................................................................... xxxvi

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

1.1 Introduction .......................................................................................................... 2 1.2 Standard Ablation Procedures for Tumor Debulking and Immunomodulation .................................................................................................................................... 3 1.3 Histotripsy as a Tumor Ablation Therapy .......................................................... 5 1.4 Animal Models for Pre-Clinical Testing of Histotripsy ..................................... 7

1.5 Outline of this Dissertation ................................................................................. 9

1.6 References ......................................................................................................... 11

Chapter 2. Histotripsy for the Treatment of Cholangiocarcinoma Liver Tumors: In Vivo Feasibility and Ex Vivo Dosimetry Study ..................................................... 22

2.1 Abstract .............................................................................................................. 23 2.2 Keywords: .......................................................................................................... 24

2.3 Introduction ........................................................................................................ 25 2.4 Procedures ......................................................................................................... 27

2.4.1 Histotripsy Systems ............................................................................ 27 2.4.2 Pressure Calibration ........................................................................... 28

2.4.3 Histotripsy In Vivo CC Ablation Procedure ......................................... 29 2.4.4 Human Liver Tumor Specimens for Ex Vivo Treatments .................... 32

2.4.5 Histotripsy Ex Vivo Ablation Procedure .............................................. 33 2.4.6 Histology & Morphological Analysis .................................................... 36

2.5 Results ................................................................................................................ 37 2.5.1 Histotripsy Ablation of Subcutaneous PDX CC Tumors ..................... 37 2.5.2 Histotripsy Bubble Cloud Generation in Ex Vivo Tissue Specimens... 39

2.5.3 Histotripsy Ablation of Ex Vivo HCC Specimens ................................ 39 2.5.4 Histotripsy Ablation of Ex Vivo CLM Specimens ................................. 40

Page 11: Determining the Oncological and Immunological Effects of

xii

2.5.5 Histotripsy Ablation of Ex Vivo CC Specimens ................................... 43 2.5.6 Histotripsy Ablation of Ex Vivo LP Specimens .................................... 44

2.6 Discussion ......................................................................................................... 46 2.7 Conclusion ......................................................................................................... 51 2.8 Acknowledgement ............................................................................................. 51 2.9 References ......................................................................................................... 53

Chapter 3. Histotripsy for Non-Invasive Ablation of Cholangiocarcinoma Carcinoma in a Patient-Derived Xenograft Model .................................................... 61

3.1 Abstract .............................................................................................................. 62 3.2 Keywords ........................................................................................................... 63 3.3 Introduction ........................................................................................................ 64 3.4 Materials and Methods ...................................................................................... 66

3.4.1 Tumor Injections and Health Monitoring ............................................. 66 3.4.2 Histotripsy Setup ................................................................................ 67

3.4.3 Histotripsy Treatment Procedures ...................................................... 68 3.4.4 Pilot Studies to Confirm Ablation Dose and Treatment Margins ......... 69

3.4.5 Survival Study ..................................................................................... 71 3.4.6 Necropsy and Histopathology ............................................................. 72

3.4.7 Statistics ............................................................................................. 73 3.5 Results ................................................................................................................ 74

3.5.1 Pilot Studies of Ablation Dose and Margins ........................................ 74

3.5.2 Survival Study ..................................................................................... 75 3.6 Discussion ......................................................................................................... 79

3.7 Conclusion ......................................................................................................... 82 3.8 Acknowledgment ............................................................................................... 83

3.9 References ......................................................................................................... 84

Chapter 4. Histotripsy Ablation in Preclinical Animal Models of Cancer and Spontaneous Tumors in Veterinary Patients ............................................................ 91

4.1 Abstract .............................................................................................................. 92 4.2 Introduction ........................................................................................................ 94 4.3 Procedures ....................................................................................................... 100

4.3.1 Histotripsy Systems and Pressure Calibrations ................................ 100 4.3.2 Hydrophone Focal Pressure Measurements, Beam Profiles, and Optical Imaging ......................................................................................... 103 4.3.3 Murine Tumor Models ....................................................................... 104 4.3.4 RAG2/IL2RG Deficient Porcine Tumor Model .................................. 106

4.3.5 Histotripsy Treatment of Porcine Pancreas and Liver ....................... 108 4.3.6 Spontaneous Tumors in Canine Patients ......................................... 110

4.3.7 Histotripsy Ablation of Spontaneous Canine Tumors ....................... 111 4.3.8 Histology & Morphological Analysis .................................................. 113

4.4 Results .............................................................................................................. 114 4.4.1 Histotripsy Systems for Small and Large Animal Studies ................. 114 4.4.2 Histotripsy Treatment of Subcutaneous Murine Tumors ................... 117

Page 12: Determining the Oncological and Immunological Effects of

xiii

4.4.3 Characterization of RAG2/IL2RG Pig Tumor Model ......................... 120 4.4.4 Histotripsy Treatment of Porcine Pancreas and Liver ....................... 124

4.4.5 Veterinary Clinical Oncology Patient Populations for Histotripsy Development ............................................................................................. 125 4.4.6 Histotripsy Ablation of Spontaneous Canine Tumors ....................... 136

4.5 Discussion ....................................................................................................... 139 4.6 Conclusion ....................................................................................................... 149

4.7 Acknowledgment ............................................................................................. 150 4.8 References ....................................................................................................... 151

Chapter 5. Establishing a SCID-Like Porcine Model of Human Cancer for Novel Therapy Development with Pancreatic Adenocarcinoma and Irreversible Electroporation .......................................................................................................... 169

5.1 Abstract ............................................................................................................ 170 5.2 Keywords ......................................................................................................... 171

5.3 Introduction ...................................................................................................... 172 5.4 Materials And Methods ................................................................................... 176

5.4.1 Generation of Immunodeficient Pigs ................................................. 176 5.4.2 Animal Care ...................................................................................... 178

5.4.3 Tumor Injection and Monitoring ........................................................ 178 5.4.4 Immunohistochemistry ...................................................................... 179 5.4.5 Ultrasound Imaging .......................................................................... 180

5.4.6 Tissue Handling and Electroporation ................................................ 180 5.5 Results .............................................................................................................. 183

5.5.1 Generation of immunodeficient pigs carrying targeted deletions in RAG2/IL2RG using the CRISPR/Cas9 system .......................................... 183

5.5.2 Panc01 tumors successfully engrafted in all pigs carrying the targeted RAG2/IL2RG deletion ................................................................................ 185

5.5.3 Panc01 tumors are histopathologically similar to Pan02 and PDX tumors generated in mice .......................................................................... 187 5.5.4 Panc01 tumors demonstrate similar electrical properties to primary patient derived pancreatic cancer tissue and healthy porcine tissues ....... 191

5.6 Discussion ....................................................................................................... 195 5.7 Abbreviations ................................................................................................... 199 5.8 Funding Sources ............................................................................................. 199 5.9 References ....................................................................................................... 200 5.10 Supplemental Figure ..................................................................................... 209

Chapter 6. Feasibility of Histotripsy Pancreas Ablation in a Large Pig Model . 210 6.1 Abstracts .......................................................................................................... 211

6.2 Keywords: ........................................................................................................ 212 6.3 Introduction ...................................................................................................... 213 6.4 Materials and Methods .................................................................................... 216

6.4.1 Animal Monitoring and Anesthesia ................................................... 216 6.4.2 Histotripsy Systems and Pressure Calibrations ................................ 218

Page 13: Determining the Oncological and Immunological Effects of

xiv

6.4.3 Hydrophone Focal Pressure Measurements and Beam Profiles ...... 219 6.4.4 Histotripsy In Vivo Pancreas Ablation Procedure ............................. 220

6.4.5 Computed Tomography Imaging ...................................................... 222 6.4.6 Necropsy & Histological Analysis ..................................................... 223

6.5 Results .............................................................................................................. 223 6.5.1 Establishing Safe, Non-Invasive Therapeutic Protocol ..................... 223 6.5.2 Physiological Outcomes ................................................................... 226

6.6 Discussions ..................................................................................................... 230 6.7 Conclusion ....................................................................................................... 232 6.8 Acknowledgements ......................................................................................... 232 6.9 References ....................................................................................................... 233

Chapter 7. Immunological Effects of Histotripsy for Cancer Therapy ............... 239

7.1 Abstract ............................................................................................................ 240 7.2 Keywords: ........................................................................................................ 241

7.3 Introduction ...................................................................................................... 242 7.4 The Role of Ablation in Immunomodulation ................................................. 243

7.4.1 The Tumor Microenvironment and Tumor Associated Immune Cells 247 7.4.2 Mechanisms for Shifting the Tumor Microenvironment to Pro-Inflammatory .............................................................................................. 252

7.5 Histotripsy: Non-Thermal, Non-Invasive Tumor Ablation ............................ 255 7.6 Immunologic Effects of Histotripsy ............................................................... 258

7.6.1 Decreases in Pro-Tumor Immune Cells ............................................ 258 7.6.2 Histotripsy Produces Damage Associated Molecular Patterns that Directly Increase Local Cellular Immune Responses ................................ 261 7.6.3 Pro-Inflammatory Cytokines and Chemokines Associated with Histotripsy Based Tumor Ablation Modalities ............................................ 264 7.6.4 Histotripsy Significantly Alters Immune Cell Populations Systemically and in the Tumor Microenvironment .......................................................... 267 7.6.5 Improved Engagement of the Adaptive Immune System and Increases in the Systemic Anti-Tumor Immune Response ........................................ 268

7.6.6 Combination Therapy with Checkpoint-Inhibitors ............................. 274

7.7 Conclusion and Future Outlook ..................................................................... 277 7.8 Acknowledgements ......................................................................................... 278 7.9 References ....................................................................................................... 279

Chapter 8. Histotripsy Ablation Alters the Tumor Microenvironment and Promotes Immune System Activation in a Subcutaneous Model of Pancreatic Cancer 299

8.1 Abstract ............................................................................................................ 300

8.2 Keywords ......................................................................................................... 301 8.3 Introduction ...................................................................................................... 302 8.4 Procedures ....................................................................................................... 304

8.4.1 Tumor Injections and Monitoring ...................................................... 304 8.4.2 Histotripsy Set Up ............................................................................. 305

Page 14: Determining the Oncological and Immunological Effects of

xv

8.4.3 n Vitro Ablations Treatment Parameters ........................................... 307 8.4.4 In Vivo Histotripsy Treatment ........................................................... 309

8.4.5 Determining DNA Release and Quality ............................................ 312 8.4.6 Determining Protein Release and Quality ......................................... 312 8.4.7 Histopathology .................................................................................. 312 8.4.8 Profiling Gene Expression and Pathway Analysis ............................ 313 8.4.9 Flow Cytometry Panel Staining......................................................... 314

8.4.10 HMGB1 Serum ELISA .................................................................... 315 8.4.11 Statistics ......................................................................................... 315

8.5 Results .............................................................................................................. 316 8.5.1 Different Ablation Modalities Show Differential Release of Damage Associated Molecular Patterns (DAMPs) and Potential Antigens .............. 316

8.5.2 Histotripsy is an Effective Tumor Ablation Modality in the Subcutaneous Pan02 Model ..................................................................... 318

8.5.3 Histotripsy Ablation Results in Increased Acute Cellular Immune Response .................................................................................................. 321

8.5.4 Histotripsy Alters Immune Cell Composition in the Tumor Microenvironment ...................................................................................... 323

8.6 Discussion ....................................................................................................... 325 8.7 Conclusion ....................................................................................................... 331 8.8 Acknowledgment ............................................................................................. 332

8.9 References ....................................................................................................... 333 8.10 Supplemental Table and Figure Legends .................................................... 342

Chapter 9. Conclusion and Future Directions ..................................................... 344 9.1 Introduction ...................................................................................................... 345

9.2 How can we safely and efficacious ablate more difficult tumors? .............. 345 9.3 Can we use a large animal model to develop histotripsy for acoustically difficult to target tumor locations? ...................................................................... 346 9.4 How do different tumor types immunologically respond to histotripsy? ... 348 9.5 Overall Conclusion .......................................................................................... 350

Appendix A. Employing Novel Porcine Models of Subcutaneous Pancreatic Cancer to Evaluate Oncological Therapies ............................................................. 351

A.1 Abstract ........................................................................................................... 352 A.2 Keywords ......................................................................................................... 352 A.3 Introduction ..................................................................................................... 353 A.4 Materials .......................................................................................................... 355

A.4.1 Generating RAG2/IL2RG Deficient Pigs .......................................... 355 A.4.2 Maintaining Cells in Culture ............................................................. 356

A.4.3 Harvesting Cells for Injection............................................................ 357 A.4.4 Sterile Passing in of the Cells........................................................... 357 A.4.5 Injecting Tumors ............................................................................... 357 A.4.6 Monitoring Tumors and Health ......................................................... 357 A.4.7 Ultrasound Imaging (see Note 6) ..................................................... 358

Page 15: Determining the Oncological and Immunological Effects of

xvi

A.4.8 Necropsy .......................................................................................... 358 A.5 Methods ........................................................................................................... 359

A.5.1 Generating RAG2/IL2RG Deficient Pigs .......................................... 359 A.5.2 Maintaining Cells in Culture ............................................................. 360 A.5.3 Harvesting Cells for Injection............................................................ 360 A.5.4 Sterile Passing in of the Cells........................................................... 361 A.5.5 Injecting Tumors ............................................................................... 362

A.5.6 Monitoring Tumors and Health ......................................................... 363 A.5.7 Ultrasound Imaging .......................................................................... 364 A.5.8 Necropsy .......................................................................................... 365

A.6 Notes ................................................................................................................ 366 A.7 References ....................................................................................................... 369

Page 16: Determining the Oncological and Immunological Effects of

xvii

List of Figures

Figure 1-1 Histotripsy is a non-invasive ablation method that uses focused ultrasound to

generate a cavitation bubble cloud, resulting in the complete destruction of targeted

tissues with high precision. .............................................................................................. 5

Figure 2-1 In Vivo Ablation Setup. (A) Histotripsy murine experiments were conducted

using a custom 1MHz small animal histotripsy system. (B) Subject’s tumors were

submerged in degassed water at the transducer focus in order for (C) histotripsy to be

applied to the tumor non-invasively guided by real-time ultrasound imaging. ............... 30

Figure 2-2 Ex Vivo Tissue Ablation Setup. Histotripsy dose experiments were conducted

using a 700kHz transducer with the focus aligned in the center of the targeted tissue,

with guidance from ultrasound imaging aligned orthogonally (A-B). Tissues were

imbedded within the center of a gelatin phantom (C). ................................................... 32

Figure 2-3 Dotted orange circles on US images (A-D) indicate approximate region of

ablation. Low magnification H&E images shows ablation margins (E-H). High

magnification H&E images of control and ablated region (J-L). Black boxes on 10x

images indicate area magnified in respective 40x images. Scale bar on 10x H&E

images = 100um (E-H), 40x scale bar = 50um (I-J). ..................................................... 38

Figure 2-4 Comparison of untreated human HCC tumors with ex vivo ablations at 500

pulses/point, 1000 pulses/point, and 2000 pulses/point. 10x magnification scale bar=

100um (A-D), 40x scale bar= 50um (E-H). Black boxes on 10x images indicate area

magnified in respective 40x images. Histology images are representative of the samples

treated. .......................................................................................................................... 40

Page 17: Determining the Oncological and Immunological Effects of

xviii

Figure 2-5 Comparison of untreated human CLM tumors with those treated at 500

pulses/point, 1000 pulses/point, 1500 pulses/point, and 2000 pulses/point. 10x

magnification scale bar= 100um (A-E), 40x scale bar= 50um (F-J). Black boxes on 10x

images indicate area magnified in respective 40x image. Histology images are

representative of the samples treated. .......................................................................... 41

Figure 2-6 Comparison of untreated human CC tumors with those treated at 1000

pulses/point, 1500 pulses/point, 2000 pulses/point, and 4000 pulses/point. 10x

magnification scale bar= 100um (A-E), 40x scale bar= 50um (F-J). Black boxes on 10x

images indicate area magnified in respective 40x image. Histology images are

representative of the samples treated. .......................................................................... 42

Figure 2-7 Supplemental Figure S1. Single sample of Cholangiocarcinoma Ablated at

4000 pulses/point illustrating fibrosis dependence to susceptibility. Region of fibrosis (A)

showed no damage, while less fibrotic region (B) had sporadic ablated foci. High

magnification of the region in B indicated by the black box is Figure 7E 4000

pulses/point location. Scale bars = 500um. * Indicate ablated region. .......................... 44

Figure 2-8 Non-steatotic liver parenchyma treatment at 250 pulses/point, 500

pulses/point, and 1000 pulses/point demonstrating less uniform ablations. 10x

magnification scale bar= 100um (A-D), 40x scale bar= 50um (E-H). Black boxes on 10x

images indicate area magnified in respective 40x images. Histology images are

representative of the samples treated. .......................................................................... 45

Figure 2-9 Steatotic liver parenchyma treatment at 250 pulses/point, 500 pulses/point,

and 1000 pulses/point demonstrating less uniform ablations. 10x magnification scale

bar= 100um (A-D), 40x scale bar= 50um (E-H). Black boxes on 10x images indicate

Page 18: Determining the Oncological and Immunological Effects of

xix

area magnified in respective 40x images. Arrows indicating example cells within an

ablated region that appear to be intact. Histology images are representative of the

samples treated. ............................................................................................................ 46

Figure 3-1 Rodent system treatment set up (A). Ultrasound imaging confirmed presence

of bubble cloud (indicated by the orange arrow) within CC tumors during treatment (B).

For treatment automation, each tumor was defined as a spheroid divided into disks (C).

Each treatment disk consisted of a series of points that were used for raster scanning

(D). White arrow in (D) represents the direction of therapeutic ultrasound wave

propagation. .................................................................................................................. 70

Figure 3-2 Schematic showing the planned ablation volumes treatments utilized for this

study. Negative-margin treatments were set to have the entire treatment contained

within the tumor; zero-margin treatments were set to treat as close to 100% of the tumor

as possible without including a significant amount of non-tumor tissue; and positive-

margin treatments were set to treat the entire tumor along with sufficient margin that

encompassed enough non-tumor tissue to ensure complete ablation of the entire tumor.

Arrows indicate tumor tissue not targeted by each planned ablation zone while * indicate

intestinal tissue within the treatment margin. Figure created using Biorender.com. ...... 72

Figure 3-3 Histological examination revealed undisturbed tumor stroma in acute

untreated mice (A) and completely acellular targeted regions in treated tumors (B).

Comparing to before treatment (C), the hypoechoic regions were identifiable in the

ultrasound imaging after treatment (D). The treatment area, represented by the dotted

yellow circle, closely matched the negative-margin planned ablation volume on

ultrasound imaging. ....................................................................................................... 73

Page 19: Determining the Oncological and Immunological Effects of

xx

Figure 3-4 Using positive, more aggressive margins (n=2) there was more significant

skin lesions and subcutaneous bruising that extended well beyond the treatment area

across the abdomen (A). Darkened, parrelel lesions that correspond to treatment disks

are emphasized with purple arrows. At necropsy, these more aggressive treatments

revealed free fluid and intestinal contents within the abdomen and a clear intestinal

perforation, indicated by the green arrow (B). ............................................................... 74

Figure 3-5 No significant skin breaks were observed during treatment (A). Immediately

after treatment, there was slight bruising surrounding the tumors, indicated by the blue

arrow (B). Three days after treatment there was no skin damage in the untreated mice

(C) and small, pitted scabs over the treated areas, noted by the red arrow (D). ........... 76

Figure 3-6 Average tumor diameter (mean + SEM) reduction after treatment, after

treatment the difference between groups was significant at every time point (* p<0.05)

(A). Individual treated and untreated mouse tumor measurements, emphasizing that

multiple treated animals reached a tumor diameter of zero (B). Vertical dotted line

emphasizes treatment on day 8. Horizontal dotted line at 14mm emphasizes maximum

tumor diameter that any individual animal was allowed to reach in this study based on

our IACUC protocols. .................................................................................................... 77

Figure 3-7 Progression free and overall survival increased (p=0.09 and 0.03

respectively) in treatment group compared to untreated. Progression free survival was

defined as surpassing 120% of the average tumor diameter the day of treatment (5.29

mm). Vertical dotted line emphasizes treatment on day 8. ............................................ 78

Figure 3-8 Histological examination revealed undisturbed tumor stroma in chronic (A)

untreated mice and tumor regrowth in chronic treated tumors (B). The liver (C-D) and

Page 20: Determining the Oncological and Immunological Effects of

xxi

lung (E-F) parenchyma in both untreated (E,C) and treated (D,F) chronic mice were

found to be benign. Scale bar = 500um. ....................................................................... 79

Figure 4-1 Example Small Animal Histotripsy System. (A) A small animal system

consisting of a 1) 1 MHz therapy transducer with an 2) imaging transducer coaxially

aligned and mounted to a 3) 3D motorized positioning system to deliver hisotripsy to

target submerged in degassed water on a 5) subject stage. A custom-built electronic

driving system is controlled with computer software. (B) Linear ultrasound imaging

probe coaxially aligned with 1 MHz therapy transducer used to (C) treat tumors in-vivo.

(D) Acoustic waveform for 1 MHz transducer collected with a hydrophone at a peak

negative pressure of ~10 MPa. (E) 1 MHz bubble cloud captured on a high-speed

camera. ....................................................................................................................... 101

Figure 4-2 Example Large Animal Histotripsy System. (A) Histotripsy system designed

for liver cancer ablation consisting of 1) therapy and 2) imaging transducers attached to

an 3) articulating arm with a 4) motorized micro-positioner controlled by 5) custom

software. (B) Image of therapy transducer with co-axially aligned imaging probe used for

(C) previous in vivo liver studies. (D) Acoustic waveform for 500 kHz transducer

collected with a hydrophone at a peak negative pressure of ~10 MPa. (E) 500 kHz

bubble captured on a high-speed camera. .................................................................. 102

Figure 4-3 Histotripsy Tumor Ablation in Murine Models. (A) Subcutaneous murine

pancreatic Pan02 tumor. (B) Representative primary human pancreatic tumor. (C)

Human CC PDX murine xenografted tumor. (D) Ultrasound images of histotripsy bubble

clouds forming in RCC, (E) 4T1, and (F) CC PDX tumors. (G-I) Histology demonstrating

full ablation within histotripsy targeted (G) RCC, (H) 4T1, and (I) CC PDX tumors. .... 118

Page 21: Determining the Oncological and Immunological Effects of

xxii

Figure 4-4 RAG2/IL2RG Immunocompromised Pig Model for Cancer Research. (A)

Example of a Pan01 (pancreatic cancer) subcutaneous tumor nodule growing behind

the ear and (B) once excised. (C) Control Matrigel plugs exhibits no growth. (D, G) U-

251 Human Glioblastoma, (E, H) 4T1 murine breast cancer, and (F, I) HepG2 human

HCC are shown by histology under low and high magnification, respectively. ............ 121

Figure 4-5 Histotripsy Treatment of Porcine Pancreas and Liver. (A) Image of the water

bowl and site preparation with fluid repellent adhesive surgical drape for liquid

containment during histotripsy. (B) Acoustic window for pancreas ultrasound treatment

identified on pre-treatment imaging. (C) Gas in the stomach and GI tract encountered

prior to histotripsy treatment. (D-E) Guided by real-time ultrasound imaging, histotripsy

generated a clearly defined bubble cloud in (D) liver and (E) pancreas. (F) Gross image

of 0.5 cm lesion in pancreas following histotripsy. (G) Off-target effects around the

stomach and GI tract. (H-I) Representative histopathology images confirm regions of

ablation in the pancreas. ............................................................................................. 123

Figure 4-6 Example Spontaneous Tumors in Canines. (A) Soft Tissue Sarcoma (STS)

presented as a firm, fixed, painful soft tissue mass at the right proximal forelimb of a

dog. (B) Contrast-enhanced CT consistent with a soft tissue attenuating and moderately

contrast enhancing mass (yellow asterisk). The mass lies lateral to the triceps muscle

and arises from the ascending pectoral muscle. (C) HIFU treatment of the tumor to

partial ablation. An Echopulse unit by Theraclion was used. (D) Gross appearance of a

cross-section of the tumor, 4 days post HIFU treatment. The yellow arrows point to the

ablated tumor volume. (E) CT image of a rostral maxillary tumor in a dog, indicated in

circled area. Note the aggressive invasion of the tumor into the maxilla, causing bone

Page 22: Determining the Oncological and Immunological Effects of

xxiii

lysis. (F) CT image of a humeral osteosarcoma in a dog, indicated by the arrow. Note

the aggressive nature of the tumor, causing bone lysis and proliferation. (G) CT image

of a liver tumor in a dog, indicated by the circle. (H) CT image of a pancreatic tumor in a

dog, indicated by the arrow. (I) CT image of a metastatic lymph node in the same

patient, indicated by a circle. ....................................................................................... 130

Figure 4-7 Histotripsy Treatment of Canine Soft Tissue Sarcoma. (A-B) Image of

spontaneous soft tissue sarcoma with estimated tumor size of ~7-8 cm in diameter. (C)

Contrast-enhanced CT collected before histotripsy treatment. (D) Histotripsy treatment

experimental set-up. (E) Real-time ultrasound image collected during histotripsy

treatment showing the bubble cloud visible as a hyperechoic, oscillating zone (arrow).

(F) Contrast-enhanced CT collected 4 days after histotripsy treatment with decreased

contrast uptake visible in the zone of ablation. (G) Gross morphological analysis of

resected canine soft tissue sarcoma. Box indicates treatment region, identified by

location and presence of tissue necrosis and hemorrhage. (H) H&E image of untreated

canine soft tissue sarcoma (20x magnification) with intact cells and undisturbed

extracellular matrix is observable. (I) H&E image of canine soft tissue sarcoma tissue

treated with histotripsy (40x magnification) showing extensive necrosis and hemorrhagic

appearance. ................................................................................................................ 136

Figure 4-8 Histotripsy Treatment of Canine Osteosarcoma. (A-B) Image of an

undisturbed spontaneous canine osteosarcoma in the left hind limb. Significant soft

tissue swelling was present, creating an estimated zone of ~30 cm of affected tissue.

(C) Contrast-enhanced CT collected before histotripsy treatment. (D) Histotripsy

treatment experimental set-up. (E) Real-time ultrasound image collected during

Page 23: Determining the Oncological and Immunological Effects of

xxiv

histotripsy treatment with the bubble cloud visible as a hyperechoic, oscillating zone

(arrow). (F) Contrast-enhanced CT collected 1 day after histotripsy treatment. .......... 137

Figure 5-1 Generation of RAG/ IL2RG knockout pigs. (A) Schematic approach for

RAG2/IL2RG knockout pig production. First, the sgRNA and Cas9 mRNA was

synthesized via in vitro transcription. Second, an optimal concentration of sgRNA and

Cas9 was injected into presumable zygotes. Third, the injected embryos were

transferred into a surrogate gilt. At the end of gestation, piglets were born via

hysterectomy and maintained in gnotobiotic isolators. (B) Summary of piglet genotypes

in IL2RG and RAG2 gene. Six piglets were born and analyzed. During PCR analysis,

RAG2 was not amplified from the Piglet #2 DNA sample, suggesting a large deletion.

Biallelic genotype indicates two differently modified alleles. Homozygous indicates one

type of modified alleles. (C) Genotype of Piglet 1 as a representation of small deletions

or insertions introduced by the CRISPR/Cas9 system. ............................................... 182

Figure 5-2 Confirmation of SCID-Like Phenotype. FACS analysis to detect the presence

of lymphocytes in RAG2/IL2RG knockout piglets. Compared to wild-type control, SCID

piglet possessed lower level of B (CD79A), T (CD3E), and NK (CD3E-/CD56+) cells. 185

Figure 5-3 Successful Engraftment of Panc01 Cells. (A) Tumor growth was measured

three times per week for each pig. All animals developed palpable tumors that

continued to grow over the course of the study. (B) Pig weight was monitored

throughout the duration of the study and utilized as a surrogate for animal morbidity. 185

Figure 5-4 Subcutaneous Tumors Generated Ample, High-Quality Pancreatic Cancer

Tissue for Subsequent Ex Vivo Tumor Ablation Assessments. (A) Tumor growth was

readily observable above the skin behind the ear where the Panc01 cells were injected.

Page 24: Determining the Oncological and Immunological Effects of

xxv

(B) No observable foci were located in regions where Matrigel alone (control) was

injected. (C) Ultrasound image confirmed the volumetric growth of the tumors. The

tumor is outlined here in white for clarity. (D-E) Necropsy of the tumors confirmed that

(D) they were confined to the dermis and (E) approximately the same size as measured

extra-dermally. ............................................................................................................ 187

Figure 5-5 Histopathology Comparison of Pre-Clinical Pancreatic Cancer Animal

Models. Human PDX, mouse Pan02, and human Panc01 were compared. (A)

Subcutaneous tumors in the mouse Pan02 model are densely cellular often with central

areas of necrosis (asterisk). Individual cells are spindle-shaped and arranged in vague,

irregular streams. (B) Subcutaneous tumors in the PDX mice are also highly cellular and

also exhibit foci of necrosis (asterisk). At higher magnification, these cells are more

polygonal and are often arranged to form irregular tubular structures with lumens

containing inflammatory cells and debris (arrows). (C) Subcutaneous tumors in the

porcine Panc01 model are also densely cellular and variably exhibit foci of necrosis.

Individual cells are more polygonal in shape and rarely attempt to recapitulate glandular

structures (arrows). ..................................................................................................... 188

Figure 5-6 Immunohistochemical Confirmation of Ductal Cells in Pre-Clinical Pancreatic

Cancer Animal Models. (A) Human PDX, (B) mouse Pan02, and (C) human Panc01

were compared for CK19 expression. Brown color indicates positive color. Nuclei are

stained in dark blue, and remaining cell features are highlighted in light blue. ............ 189

Figure 5-7 Primary human, SCID-like Panc01, and murine PDX tumors demonstrate

similar conductivity increases following high voltage pulsed electric field. Ex vivo tissue

samples directly from patients, RAG2/IL2RG animals, or NSG mice were sectioned and

Page 25: Determining the Oncological and Immunological Effects of

xxvi

fit within a PDMS mold to impose a cylindrical shape factor. After sample preparation,

individual samples were exposed to IRE pulses with varying electric field magnitudes.

A) Summary of initial conductivities calculated from normal and cancerous SCID-like

(black), human (red), and murine PDX (gray) tissue. B) The conductivity for each tissue

type increased with the applied electric field. C) Percent increase in tissue conductivity

at varying fields was determined from the sample-specific pre-pulse values. D) The

adjusted conductivity is calculated from the percent difference values; here, the

conductivities are normalized to the average initial tissue conductivity and adjusted

based on the percent change. ..................................................................................... 191

Figure 5-8 Impedance for healthy tissue excised from the RAG2/IL2RG pigs correlate

adult swine and humans. A Gamry Reference 600 potentiostat (Gamry, Warminster,

PA, US) was used to record tissue impedance within a frequency range of 103 to 106

Hz. The real part of the tissue impedance was used in the calculation of tissue

conductivity for further comparison to values previously reported in literature. A)

Comparison of electrical conductivity from SCID-like pig tissue to healthy porcine

(pancreas/brain) or human (prostate) tissue. The B) pancreatic, C) brain, and D)

prostate tissue demonstrate similar electrical behavior to their normal counterparts

across a wide frequency range. Each data point (dot) represents reported values for

normal tissue conductivity, while the black line represents data gathered in this study

and the red line represents normal porcine tissue. (*p < 0.05). ................................... 192

Figure 5-9 Supplemental Figure 5.S1. Genotyping of piglets carrying modified IL2RG

and RAG2 gene. Six piglets were born and analyzed. During PCR analysis, RAG2 was

Page 26: Determining the Oncological and Immunological Effects of

xxvii

not amplified from the Piglet #2 DNA sample, suggesting a large deletion. No wild-type

allele was identified. .................................................................................................... 209

Figure 6-1Study timeline demonstrating the relative time for treatment, custard feeding,

blood draws, and CT imaging. ..................................................................................... 217

Figure 6-2 HistoSonics cart with 500 kHz transducer and coaxial US imaging probe

mounted onto a micropositioner (A). Freehand imaging by radiologist to identify acoustic

window (B). UMC bowl with therapy transducer submerged via adjustable arm (C). .. 218

Figure 6-3 Bubble cloud formed in liver on US imaging from HistoSonics system (pig 1,

8-6)(A). No bubble cloud visualized on US imaging and did not clearly identify the

pancreas without pudding (pig 6, 8-13)(B). Bubble cloud generated inside pancreas

where pig was fed pudding prior to histotripsy treatment (28-53) (C). ......................... 224

Figure 6-4 Intestinal gas interference with treatment prevented ideal imaging quality and

bubble formation during treatment leading to pre-focal intestinal bruising (A-B) more

highly concentrated in the more gaseous large intestine, indicated by the green arrow,

and near the targeted region, indicated by the blue arrows. Post treatment H&E

histopathology indicated that from a sample of the bruised colon, the muscularis was

not damaged but there was a large amount of ablation within the submucosa (C-D). 225

Figure 6-5 US imaging of pancreas before histotripsy (A) and after treatment with clear

hypoechoic region identifying the targeted ablated volume (B). Dotted red circle

indicated the approximate region treated .................................................................... 226

Figure 6-6 With laxative, simethicone, and custard pretreatment protocol, there was

signifcatly less bruising in preforcal bowel (A) indicated by the green arrow. Bruising

Page 27: Determining the Oncological and Immunological Effects of

xxviii

around the treatment area was more concentrated than on the bowel, but was still less

intense than the acute group (B), bruising indicated by blue arrow. ............................ 227

Figure 6-7 CT taken in the dorsal recumbent position (A) one week post treatment

shows no free fluid in the peritoneal cavity (B) ............................................................ 228

Figure 6-8 White blood cell, amylase, and lipase serum levels for 1-week survival

animals. ....................................................................................................................... 228

Figure 6-9 1-week post treatment there was no signs of morbidity within the pancreatic

tissue. Images representative of two animals. ............................................................. 229

Figure 6-10 Necropsy reviled no gross damage within the abdomen in situ (A-B). When

excised in tota no damage was found on the exterior of the liver (C-D), the spleen (E),

nor the pancreas (F-G). ............................................................................................... 229

Figure 7-1 Focal Treatment of Targeted Tumor and Mechanism for Systemic Tumor

Control. The right diagram demonstrates the principal of the abscopal effect, where the

treatment of one tumor can cause other tumors in the body to shrink or be eliminated to

varying degrees due to immune system engagement. The left diagram depicts the

simplified mechanism for achieving a systemic effect from focal therapies. DC: Dendritic

cell. MHC: Major Histocompatibility Complex. TCR: T cell receptor. ........................... 245

Figure 7-2 Pro-Tumor Immune Cells. CD66, CD66b, and CD15 are human specific

markers. Gr-1 represents both Ly6C and Ly6G, therefore does not distinguish the

MDSC subtypes, MDSCs are monocytic when Ly6C+ and granulocytic when Ly6G+.

Most of the markers available for pro-tumor immune cells are surface receptors and

ligands, exceptions listed here include the intracellular protein Arg1 and the transcription

factor FOXP3. ............................................................................................................. 249

Page 28: Determining the Oncological and Immunological Effects of

xxix

Figure 7-3 The Role of Check Point Inhibitors. Antigen Presenting Cells (APC). ........ 253

Figure 7-4 Histotripsy Schematic. Therapeutic ultrasound transducer positioned outside

of the body, focuses ultrasound waves within a targeted tissue generating a cavitation

bubble cloud. ............................................................................................................... 256

Figure 7-5 Schematic of Histotripsy’s Postulated Roles in Immune System Modulation.

Immune processes, cells, and molecules that have been shown to be modulated by

focused ultrasound therapies after mechanical ablation are summarized based upon

immune system functions: pro-tumor immune cells, cellular immunity, and systemic

immunity. ..................................................................................................................... 259

Figure 8-1 In Vivo Experimental Setup. Murine histotripsy experiments were conducted

using a 1Mhz transducer (A). Timing for treatment, euthanasia, and data collection were

set as diagramed (B). Days with histology collection are noted by cassettes, flash frozen

tumors noted by tubes with tumors, serum noted by tubes with serum, and flow

cytometry is noted by flow charts. Tumors located with ultrasound imaging were used

for planning automated ablation disks (C) and raster scan plots (D). Therapy was

guided by co-axially aligned ultrasound imaging (E). Red arrow indicates a bubble cloud

that was generated during treatment. .......................................................................... 306

Figure 8-2 Treatment flow and dosages for various ablation modalities (A). Histotripsy

experiments on cell suspensions were conducted using a 1Mhz transducer (B). ....... 308

Figure 8-3 Cell suspension ablations resulted in partial and full ablations (A). DNA

release was quantified (B) and visualized (C). Protein release was quantified (D) and

the relative release of the antigen HA was quantified from western blot bands (E).

Lowercase letters on top of bars indicate significance; bars with the sample letter

Page 29: Determining the Oncological and Immunological Effects of

xxx

designation are not significant while those that do not share a letter are significant

(p<0.05). ...................................................................................................................... 316

Figure 8-4 Ultrasound images of tumor before (A) and after (B) treatment, exhibiting

more hypoechoic central region post-histotripsy. Orange shapes on ultrasound indicate

location of tumors. Green circles indicate location of bubble cloud determined in water

prior to treatment, and were utilized for treatment planning. Histology to tumors without

(C) and with (D) treatment show decreased cellular detail after treatment. Dotted black

line outlines the necrotic core of the tissue. Scale bar on H&E images = 500 um. ...... 319

Figure 8-5 After treatment of tumors on day 8, as indicated by the black arrow, the

reduction of tumor volume was indicated by caliper measurements (A). Changes health

observed in tumor-progression free survival (B) and general survival (C). .................. 320

Figure 8-6 Analysis of mRNA expression in treated tumors showed regulation of

immune pathways (A). Regulated pathways interact with each other and are up

regulated in the acute group and downregulated in the chronic group (B). Serum

HMBG-1 levels (C) correlate with the mRNA upregulation of HMGB-1 associated

pathways. .................................................................................................................... 321

Figure 8-7 Single cell suspension from treated and untreated tumors were collected at

24 hours, 7 days, and 14 days after treatment and were stained as described for flow

cytometry to identify innate immune cells. Percentage of each innate immune cell

population analyzed was calculated as part of the total CD45+ cells stained. Example

flow cytometry plots from treated and untreated tumors at 14 days after treatment. ... 324

Figure 8-8 Single cell suspension from treated and untreated tumors were collected at

24 hours, 7 days, and 14 days after treatment and were stained as described for flow

Page 30: Determining the Oncological and Immunological Effects of

xxxi

cytometry to identify adaptive immune cells. Percentage of each adaptive immune cell

population analyzed was calculated as part of the total viable cells stained (gated as

singlets). The ratio of CD4+/CD8+ T cells was calculated as a simple ratio. Example

flow cytometry plots from treated and untreated tumors at 14 days after treatment. ... 325

Figure 8-9 Supplemental Figure 1: Whole western blot images analyzing HA antigen

released in ablated cell supernatants. ......................................................................... 342

Figure 0-1: Creating “Tent” to Ensure Subcutaneous Injection. By pulling the pig’s ear

forward, a small tent of skin will pull up forming a small tent. To ensure a subcutaneous

injection, have the bevel of the needle enter “tent” following a path indicated ............. 362

Figure 0-2: Measuring Subcutaneous Tumor Growth in Gnotobiotic Porcine Isolators.

(A) Subcutaneous tumors (indicated by arrow) grown behind the ear can be easily

viewed within two days post injection and (B) measured with plastic Vernier calipers. (C)

The .............................................................................................................................. 363

Page 31: Determining the Oncological and Immunological Effects of

xxxii

List of Tables

Table 2-1 Ex Vivo sample numbers by tissue type and Dose --------------------------------- 36

Table 5-1 Target sequences of RAG2 and IL2RG sgRNAs. ---------------------------------- 176

Table 5-2 Primers used to amplify sgRNA target regions. ------------------------------------ 178

Table 8-1 Subject Numbers Per Experimental Group ------------------------------------------ 309

Table 8-2 Flow Cytometry Immune Cell Markers ------------------------------------------------ 313

Table 8-3 Supplemental Table 1: Genes analyzed from SuperArry rtPCR and calculated

fold regulation. -------------------------------------------------------------------------------------------- 343

Page 32: Determining the Oncological and Immunological Effects of

xxxiii

List of Abbreviations

ACCRC: Animal Cancer Clinic & Research Center

ANOVA: Analysis of Variance

APC: Antigen Presenting Cell

ATCC: American Type Culture Collection

BH: Boiling Histotripsy

CBC: Complete Blood Count

CC: Cholangiocarcinoma

CCH: Cavitation Cloud Histotripsy

CLM: Colorectal Liver Metastasis

COTC: Comparative Oncology Trials Consortium

CRISPR: Clusters of Regularly Interspaced Short Palindromic Repeats

CT: Computed Tomography (imaging)

DAMP: Damage Associated Molecular Pattern

DC: Dendritic Cell

DNA: Deoxyribose Nucleic Acid

ERF: Epidermal Growth Factor

FOPH: Fiber Optic Probe Hydrophone

FPGA: Field Programmable Gate Array

FWHM: Full width Half Maximum

H&E: Hematoxylin and Eosin

HA: Hemagglutinin

HCC: Hepatocellular Carcinoma

Page 33: Determining the Oncological and Immunological Effects of

xxxiv

HIFU: High Intensity Focused Ultrasound

IACUC: Institutional Animal Care and Use Committee

IL: Interleukin

IRB: Institutional Review Board

IRE: Irreversible Electroporation

MDSC: Myeloid Derived Suppressor Cell

MHC: Major Histocompatibility Complex

mHIFU: Mechanical High Intensity Focused Ultrasound

MRI: Magnetitic Resonance Imaging

NCI: National Cancer Institute

NK: Natural Killer (cell)

NSG: Nod-SCID Gamma

OCM: Oncopig Cancer Model

OS: Osteosarcoma

PDX: Patient Derived Xenograft

PRF: Pulse Repetition Frequency

RCC: Renal Cell Carcinoma

RFA: Radiofrequency Ablation

SCID: Severe Combined Immunodeficiency

SD: Standard Deviation

SEM: Standard Error of the Mean

STS: Soft Tissue Sarcoma

TAM: Tumor Associated Macrophage

Page 34: Determining the Oncological and Immunological Effects of

xxxv

TDLN: Tumor Draining Lymph Node

tHIFU: Thermal High Intensity Focused Ultrasound

TKX: Telazol, Ketamine, Xylazine

TME: Tumor Microenvironment

US: Ultrasound

VCRO: Veterinary Clinical Trials Network and Clinical Research Office

VMCVM: Virginia-Maryland College of Veterinary Medicine

Page 35: Determining the Oncological and Immunological Effects of

xxxvi

Attributions for Co-Authored Papers

Alex Simon: Assisted with experimentation, manuscript preparation, and manuscript

review of Chapters 2, 3, 4, 7, and 9.

Allison Zeher: Assisted with experimentation and manuscript review of Chapter 9.

Christopher Bryon, DVM: Assisted with manuscript preparation and manuscript review

for Chapter 4.

David Luyimbazi, MD: Assisted with experiment planning and manuscript review for

Chapters 2, 3, and 7.

Douglas Grider, MD: Provided pathology review for data collection and manuscript

review to Chapters 2, 3, and 7.

Eli Vlaisavljevich, PhD: Assisted with experiment planning, manuscript preparation,

and manuscript review for Chapters 2-9.

Hannah Sheppard: Assisted with experimentation and manuscript review of Chapter 7.

Holly Morrison: Assisted with experimentation and manuscript review of Chapter 6.

Irving C Allen, PhD: Assisted with experiment planning, manuscript preparation, and

manuscript review for Chapters 2-9.

Jaqueline Sereno: Assisted with experimentation and manuscript review of Chapter 9.

Jessica Gannon: Assisted with experimentation, manuscript preparation, and

manuscript review of Chapters 4, 6, 7, and 10. Co-first author of the submitted

manuscripts associated with Chapters 4 and 7.

Joan Vidal-Jove, MD, PhD: Assisted with experiment planning and manuscript review

for Chapters 2 and 7.

Page 36: Determining the Oncological and Immunological Effects of

xxxvii

Joanne Tuohy, DVM, PhD: Assisted with experiment planning, manuscript preparation,

and manuscript review for Chapters 4.

John H Rossmeisl, DVM: Assisted with manuscript preparation and manuscript review

for Chapter 4.

Kayla Farrell: Assisted with experimentation and manuscript review of Chapter 6.

Kenneth N Aycock: Assisted with experimentation and manuscript review of Chapter 6.

Khan Mohammad Imran: Assisted with experimentation, manuscript preparation, and

manuscript review of Chapters 9.

Kiho Lee, PhD: Assisted with experiment planning, manuscript preparation, and

manuscript review for Chapters 5 and 6.

Kyungjun Uh: Assisted with experimentation, manuscript preparation, and manuscript

review of Chapters 5 and 6.

Lauren Arnold: Assisted with experimentation, manuscript preparation, and manuscript

review of Chapters 4 and 7. Co-first author of the submitted manuscript associated with

Chapter 4.

Margaret Nagai-Singer: Assisted with experimentation, manuscript preparation, and

manuscript review of Chapters 4-7.

Martha M Larson, DVM: Provided radiology assistance for experimentation and

manuscript review of Chapter 7.

Melvin Lorenzo: Assisted with experimentation and manuscript review of Chapter 6.

Michael Edwards, DVM, PhD: Provided radiology assistance for experimentation and

manuscript review of Chapter 7.

Page 37: Determining the Oncological and Immunological Effects of

xxxviii

Mishal Mendiratta-Lala, MD: Assisted with experiment planning and manuscript review

of Chapter 7.

Natalie Beitel-White: Assisted with experimentation, manuscript preparation, and

manuscript review of Chapters 6 and 9.

Neha Singh: Assisted with experimentation, manuscript preparation, and manuscript

review of Chapters 4 and 7.

Nikolaos Dervisis, DVM: Assisted with experiment planning, manuscript writing, and

manuscript review for Chapter 4.

Peter Weber: Assisted with experimentation and manuscript review of Chapter 2.

Rafael Davalos, PhD: Assisted with experiment planning and manuscript review for

Chapter 6 and 9.

Rebecca Brock, PhD: Assisted with experimentation and manuscript review of

Chapters 6 and 9.

Ruby Hutchison: Assisted with manuscript preparation and review of Chapter 8.

Shawna Klahn, DVM: Assisted with experiment planning, manuscript preparation, and

manuscript review for Chapter 4.

Sherrie Clark-Deener, DVM, PhD: Assisted with experimentation, manuscript

preparation, and manuscript review for Chapters 5 and 6.

Sheryl Coutermarsh-Ott, DVM, PhD: Provided pathology review for data collection

and manuscript review to Chapters 2, 3, 4, 6, 7, and 9.

Sofie Saunier: Assisted with experimentation, manuscript preparation, and manuscript

review of Chapters 2 and 3.

Page 38: Determining the Oncological and Immunological Effects of

1

Chapter 1. Introduction

Page 39: Determining the Oncological and Immunological Effects of

2

1.1 Introduction

Cancer is the second leading cause of death world-wide, with multiple tumor types

still facing abysmal 10% or lower 5-year overall survival rates. There are many reasons

attributed to this, but regardless, there is a pressing need to improve treatment options.

The core pillars of cancer therapy are chemotherapy, surgery, radiation,

immunotherapy, and ablation. The most common treatment option for solid tumors

remains surgical resection; however, many patients are not candidates due to tumor

size, location, or disease progression [1-4].

Two cancers that epitomize these problems are pancreatic cancer and

cholangiocarcinoma (CC), a cancer of the bile ducts. Pancreatic cancer is the fourth

leading cause of cancer-related deaths, with a 9% survival rate due to its late diagnosis

and lack of curative treatment options [5, 6]. Although less common, CC also has a low

survival rate of 5-10% [7]. For these malignancies, the standard treatment options are

limited to surgery, chemotherapy, and radiation [7-10]. While surgical resection is the

most curative option for these tumors, many patients do not qualify due to tumor

burden, location, or comorbid disease [2, 11, 12]. Only 20% of patients have pancreatic

tumors that can be surgically removed, and of those patients the cure rate is less than

25% [13]. Only 15% of intrahepatic CC tumors and 56% of distal CC tumors on average

are deemed resectable [14]. For patients with surgically resectable intrahepatic CC, the

5-year survival rate is still only 22-30% [9, 15] This indicates that even with the most

ideal presentation CC still has a low survival rate. In order to address the limitations that

Page 40: Determining the Oncological and Immunological Effects of

3

are faced in treating these tumor types, there has been recent development of new

ablation therapies and investigation into the potential of immunomodulation to better

fight systemic disease.

1.2 Standard Ablation Procedures for Tumor Debulking and Immunomodulation

Ablation procedures can act as a replacement for or as an adjuvant to surgery. The

most commonly used ablation modalities for treating cancers in abdominal organs are

radiofrequency ablation (RFA), microwave ablation, high-intensity focused ultrasound

(HIFU), cryoablation, and irreversible electroporation (IRE). Of these modalities, RFA,

microwave ablation, and HIFU all utilize thermal mechanisms. RFA is a minimally-

invasive ablation modality that utilizes high-frequency alternating currents to thermally

induce thermal necrosis [16]. Microwave ablation, also minimally-invasive, induces cell

death through electromagnetic microwaves that produce friction and heat [17]. HIFU, a

non-invasive focused ultrasound, ablates cells by depositing ultrasound energy at a

focal point to rapidly increase tissue temperature [18]. While these therapies have

promise, reliance on thermal mechanisms leads to inherent limitations due to the heat

sink effect [19-21] where treatments near vasculature are reduced in efficacy due to the

flow of blood through the treatment region minimizing the efficacy of heat deposition.

This also leads to non-uniform heat dissipation associated with hypervascularization

[22]. An additional limitation to these therapies is the lack of real-time treatment

monitoring, although treatment can be evaluated for success with post-treatment CT

and MR imaging.

Page 41: Determining the Oncological and Immunological Effects of

4

Alternatively, cryoablation and IRE do not rely on heat dissipation for their

mechanism of ablation. Cryoablation results in tumor cell destruction through ice-crystal

formation as a result of liquid nitrogen or argon gas delivered to the tissue, which can be

monitored in real time with medical imaging [23]. IRE utilizes short, high-voltage

electrical pulses that non-thermally open micro-pores in cell plasma membranes,

inducing cell death, and can be monitored in real-time for changes in tissue electrical

conductivity [24, 25]. Recent advancements have established that non-thermal

therapies have the potential to treat difficult tumors such as pancreatic cancer. For

example, clinical trials have shown that IRE can ablate pancreatic tumors without

damaging nearby critical structures and have had dramatic, positive effects on patient

survival that may be due to the induction of immunomodulatory mechanisms [26-31].

Given cryoablation and IRE’s non-thermal mechanisms, these procedures have been

found to release immunostimulatory molecules and lead to a more immunologically

active tumor microenvironment after treatment [16, 32]. Furthermore, recent studies

comparing the thermal ablation modalities, cryoablation, and IRE have found that the

non-thermal modalities are more potent at stimulating an anti-tumor microenvironment

[33]. However, these procedures still require patients be healthy enough, through co-

morbidities and metastatic disease, for surgery and involve surgical incisions that

increase the possibility of surgery-related injury and infection, including the minimally

invasive percutaneous procedures.

Page 42: Determining the Oncological and Immunological Effects of

5

1.3 Histotripsy as a Tumor Ablation Therapy

Histotripsy is an emerging non-invasive, non-thermal, image-guided cancer ablation

modality that has recently been approved for its first clinical trial in the United States

(NCT04573881). Histotripsy is a focused ultrasound therapy that utilizes microsecond

through millisecond pulsing regimens to generate controlled cavitation bubble clouds at

the focus, leading to precise ablation of targeted tissues [34] (Fig.1). The rapid

expansion and collapse of cavitation bubbles ablate tissues into acellular debris [35-37].

This process is well

established and is due to the

mechanical nature of the

cavitation-based ablation.

Given the mechanical

mechanism, there is tissue selectivity associated with cavitation ablation. For example,

stronger tissues, with a higher Young’s Modulus, such as vasculature and collecting

ducts are more resistant to damage [38-42]. By utilizing a fully mechanical mechanism,

tissue strength is strongly correlative with dose and susceptibility [43-45]. Studies have

shown that histotripsy is capable of ablating tissues that have a lower concentration of

collagen within the extracellular matrix [39]. This finding, in combination with the

established tissue strength-susceptibility effect, implies increased safety of treating

tumors that frequently have disorganized extracellular matrix components and are

located near stiffer critical structures, such as blood vessels or bile ducts.

Figure 1-1 Histotripsy is a non-invasive ablation method that uses focused ultrasound to generate a cavitation bubble cloud, resulting in the complete destruction of targeted tissues with high precision.

Page 43: Determining the Oncological and Immunological Effects of

6

Unlike the vast majority of other ablation therapies, histotripsy is non-thermal, and it

is not affected by the heat-sink effect, and ,therefore, remains safe and efficacious for

use near the vasculature [46, 47]. Additional benefits of histotripsy include real-time

imaging feedback with standard imaging (i.e. ultrasound, MRI, CT), highly precise-

millimeter accuracy, and the ability to treat tumors of arbitrary sizes and shapes [40, 48].

An additional benefit of histotripsy is the improved wound healing of ablation zones

compared to other ablation modalities. Histotripsy ablation of healthy rat livers showed

rapid involution of treated volumes, granulation, and growth of healthy hepatocytes

within 28 days with minimal scarring [36]. This healing process is more rapid than what

has been reported for other ablation modalities, such as RFA in humans which has

been reported as “thermal fixation” [49], where the necrotic mass is still present 14

months post-treatment due to thermal denaturation of the tissue leaving it resistant to

being broken down by normal pathways.

In mouse models, histotripsy ablation of hepatic, renal, neuroblastoma, and

melanoma tumors has resulted in significantly increased survival [50-53]. In addition to

debulking tumors, recent evidence has suggested that histotripsy has the capability to

induce a systemic immune response, as evidenced by the attenuation of metastasis and

an improvement in local and distant disease with combination immunotherapy [52, 54,

55]. This effect was shown not only in single animal-tumor treatments, but also showed

abscopal-like decreases in contralateral tumor growth in a separate untreated tumor.

This effect was found to be modest but statistically significant with histotripsy ablation

compared to the insignificant trend in mice treated with irradiation and RFA [52].

Primary studies focused on the use of histotripsy in treating tumors in model organisms

Page 44: Determining the Oncological and Immunological Effects of

7

have shown that it can be safe and effective; however, there is still plenty of work to be

done before establishing that this therapy will be efficacious in humans [50, 56, 57].

Despite the promise of histotripsy and the many positive results reported in

preclinical studies to date, only a single human clinical trial has been conducted to test

histotripsy for the treatment of cancer [58]. This recent Phase I clinical trial was

conducted with non-curative, palliative intent in patients with multifocal primary and

metastatic liver tumors, with results showing histotripsy could safely target and ablate

targeted liver tumors [58]. To our knowledge, no other human clinical trials of histotripsy

for the treatment of cancer have been conducted.

1.4 Animal Models for Pre-Clinical Testing of Histotripsy

Similar to other focal tumor ablation modalities, one of the primary factors limiting the

rapid translation of histotripsy into the clinic has been the lack of appropriate animal

models for studying the safety, effectiveness, and optimization for the treatment of a

variety of cancer types. Previous efforts to develop histotripsy for specific oncology

applications have been split into separate studies testing human-scale and miniaturized

rodent-scale systems.

Studies testing human-scale histotripsy systems for ablating tissues is normally

conducted in healthy, large animal models that have similar anatomy and physiology to

human patients, primarily pigs [48, 59-61]. For example, the same tumor ablation

systems can typically be utilized, similar clinical/surgical techniques can be refined, and

many large animal models accurately recapitulate human anatomical and physiological

Page 45: Determining the Oncological and Immunological Effects of

8

features. This is critical for imaging and ultrasound-based modalities such as histotripsy.

Although early pre-clinical studies utilizing pigs has focused on healthy animals, the

development of novel cancer porcine models is expanding the potential for pre-clinical

work. With the rise of technologies capable of inducing genetic modifications in large

mammals, such as CRISPRs and TALONs, there is an interest in the pig as a

spontaneous cancer model [62-64]. As an alternative strategy, research teams have

focused on immunocompromised pig models, where deletions in the RAG2 and IL2RG

genes have generated animals with similar characteristics to NOD scid gamma (NSG)

mice, which are susceptible to human tissue engraftment [65]. Similar to NSG mice,

human tumor cell lines and patient-derived xenograft (PDX) tissue can be engrafted [66,

67]. This allows a highly predictable tumor that can be surgically placed subcutaneously

or orthotopically in a human-sized animal with similar anatomy and physiology. In

general, pigs have yet to play a significant role in experimental oncology [62].

These large animal debulking studies are typically complemented by separately

testing histotripsy tumor ablation in rodents (mice, rats, and rabbits) using customized

small animal devices that allow histotripsy to be applied in this setting. Albeit, these

studies do not use the systems or acoustic parameters that will ultimately be developed

for use in humans in order to more rapidly investigate the biological and immunological

effects of ablation [52, 54, 56, 68]. Murine models are the dominant in vivo model in the

biomedical engineering and cancer biology fields and are essential in acquiring

fundamental data. In typical mouse models used to study tumor ablation modalities, the

vast majority of studies are mouse or human cell line based. While these models offer

excellent reproducibility and standardization, they rarely recapitulate the clinical, in situ,

Page 46: Determining the Oncological and Immunological Effects of

9

microenvironment found in patients or critical biological hallmarks of the tumor of origin.

Likewise, the lack of translational acoustic windows, limited imaging, lack of precision,

and lack of physiological relevance are all major limitations of small animal models in

the development of histotripsy and other focused ultrasound modalities.

Beyond induced tumor models, veterinary cancer patients offer unique

translational research opportunities for cancer imaging and medical device

development. Similar to humans, companion animals are genetically diverse and are

exposed to similar environments. Comparative oncology studies have revealed

significant similarities between many canine and human cancers, and over the past two

decades canines have been used for an increasing number of pre-clinical pharmacology

studies [69-77]. The spontaneous nature of canine cancers, their biological similarities

to human cancers, and the anatomic similarities between dogs and people offer highly

relevant clinical conditions for device development.

1.5 Outline of this Dissertation

This dissertation consists of three parts. Part One of this dissertation (Chapters

2-3) focuses on determining the safety and dosages for using histotripsy to treat

stronger hepatopancreatobiliary tumors using excised tissues and patient derived

xenograft (PDX) mice. Chapter 2 focuses on dosimetry studies of CC tumors, to

determine the feasibility of using histotripsy to treat dense, highly fibrotic tumors. For

Chapter 4, PDX mice bearing CC tumors were treated with histotripsy and survived to

determine the feasibility of a full ablation without debilitating off target damage.

Page 47: Determining the Oncological and Immunological Effects of

10

Part Two (Chapters 4-7) moves onto more complex models, using large animals

and veterinary patients to establish application protocols for additional tumor types.

Chapter 5 demonstrates the pros and cons of utilizing histotripsy to treat a wide range of

tumors in murine and porcine models as well as veterinary canine patients. Chapter 6

introduces an immunocompromised porcine model that can support the growth of

human tumors. Chapter 7 utilizes the newly established xenograft porcine model to

grow human pancreatic tumors and compares the physiology of these tumors and pigs

to humans, with a pilot study showing the efficacy of using these tumors for IRE

ablation. In Chapter 8, healthy porcine pancreases are ablated in vivo to determine the

safety and feasibility of using histotripsy to treat targets within the pancreas.

Part Three (Chapters 8-9) shifts to utilizing immunocompetent mouse models to

investigate the immunological effects of histotripsy in pancreatic adenocarcinoma. In

Chapter 9, the current body of literature on histotripsy’s role in modulating the immune

system locally and systemically is reviewed. Chapter 10 uses the Pan02 pancreatic

adenocarcinoma subcutaneous model in C57/Bl6 mice to investigate the release of

damage associate molecular patterns and potential antigens by histotripsy compared to

established ablation modalities as well as the shift in the local tumor microenvironment.

Page 48: Determining the Oncological and Immunological Effects of

11

1.6 References

[1] A. S. Befeler and A. M. Di Bisceglie, "Hepatocellular carcinoma: diagnosis and

treatment," Gastroenterology, vol. 122, no. 6, pp. 1609-1619, 2002.

[2] E. G. Chiorean and A. L. Coveler, "Pancreatic cancer: optimizing treatment

options, new, and emerging targeted therapies," Drug design, development and

therapy, vol. 9, pp. 3529-45, 2015, doi: 10.2147/DDDT.S60328.

[3] C. Armengol, M. R. Sarrias, and M. Sala, "Hepatocellular carcinoma: present and

future," Medicina Clínica (English Edition), vol. 150, no. 10, pp. 390-397, 2018.

[4] J. J. Hsieh et al., "Renal cell carcinoma," Nature reviews Disease primers, vol. 3,

p. 17009, 2017.

[5] R. L. Siegel, K. D. Miller, and A. Jemal, "Cancer statistics, 2017," CA: a cancer

journal for clinicians, vol. 67, no. 1, pp. 7-30, 2017.

[6] R. L. Siegel, K. D. Miller, and A. Jemal, "Cancer statistics, 2020," CA: a cancer

journal for clinicians, vol. 70, no. 1, pp. 7-30, 2020.

[7] E. U. Cidon, "Resectable cholangiocarcinoma: reviewing the role of adjuvant

strategies," Clinical Medicine Insights: Oncology, vol. 10, p. CMO. S32821, 2016.

[8] H. Manuel, "Pancreatic cancer," New England Journal of Medicine, vol. 362, no.

17, 2010.

[9] M. N. Mavros, K. P. Economopoulos, V. G. Alexiou, and T. M. Pawlik, "Treatment

and prognosis for patients with intrahepatic cholangiocarcinoma: systematic

review and meta-analysis," JAMA surgery, vol. 149, no. 6, pp. 565-574, 2014.

Page 49: Determining the Oncological and Immunological Effects of

12

[10] J. Liu et al., "Prognostic Factors and Treatment Strategies for Intrahepatic

Cholangiocarcinoma from 2004 to 2013: Population-Based SEER Analysis,"

Transl Oncol, vol. 12, no. 11, pp. 1496-1503, Nov 2019, doi:

10.1016/j.tranon.2019.05.020.

[11] M. C. Jansen, R. van Hillegersberg, R. A. Chamuleau, O. M. van Delden, D. J.

Gouma, and T. M. van Gulik, "Outcome of regional and local ablative therapies

for hepatocellular carcinoma: a collective review," Eur J Surg Oncol, vol. 31, no.

4, pp. 331-47, May 2005, doi: 10.1016/j.ejso.2004.10.011.

[12] L. Rossi et al., "Current approach in the treatment of hepatocellular carcinoma,"

World J Gastrointest Oncol, vol. 2, no. 9, pp. 348-59, Sep 15 2010, doi:

10.4251/wjgo.v2.i9.348.

[13] S. Gillen, T. Schuster, C. M. Zum Büschenfelde, H. Friess, and J. Kleeff,

"Preoperative/neoadjuvant therapy in pancreatic cancer: a systematic review and

meta-analysis of response and resection percentages," PLoS medicine, vol. 7,

no. 4, 2010.

[14] D. M. Nagorney, J. H. Donohue, M. B. Farnell, C. D. Schleck, and D. M. Ilstrup,

"Outcomes after curative resections of cholangiocarcinoma," Archives of surgery,

vol. 128, no. 8, pp. 871-878, 1993.

[15] G. Spolverato et al., "Can hepatic resection provide a long‐term cure for patients

with intrahepatic cholangiocarcinoma?," Cancer, vol. 121, no. 22, pp. 3998-4006,

2015.

Page 50: Determining the Oncological and Immunological Effects of

13

[16] O. Kepp, A. Marabelle, L. Zitvogel, and G. Kroemer, "Oncolysis without viruses—

inducing systemic anticancer immune responses with local therapies," Nature

Reviews Clinical Oncology, pp. 1-16, 2019.

[17] C. J. Simon, D. E. Dupuy, and W. W. Mayo-Smith, "Microwave ablation:

principles and applications," Radiographics, vol. 25, no. suppl_1, pp. S69-S83,

2005.

[18] Y. Keisari, "Tumor abolition and antitumor immunostimulation by physico-

chemical tumor ablation," Front Biosci (Landmark Ed), vol. 22, pp. 310-47, 2017.

[19] S. A. Curley, "Radiofrequency ablation of malignant liver tumors," The oncologist,

vol. 6, no. 1, pp. 14-23, 2001.

[20] D. S. Lu et al., "Influence of large peritumoral vessels on outcome of

radiofrequency ablation of liver tumors," Journal of vascular and interventional

radiology, vol. 14, no. 10, pp. 1267-1274, 2003.

[21] J. A. Marrero and S. Pelletier, "Hepatocellular carcinoma," Clin Liver Dis, vol. 10,

no. 2, pp. 339-51, ix, May 2006, doi: 10.1016/j.cld.2006.05.012.

[22] T. Livraghi, L. Solbiati, M. F. Meloni, G. S. Gazelle, E. F. Halpern, and S. N.

Goldberg, "Treatment of focal liver tumors with percutaneous radio-frequency

ablation: complications encountered in a multicenter study," Radiology, vol. 226,

no. 2, pp. 441-451, 2003.

[23] J. P. Erinjeri and T. W. Clark, "Cryoablation: mechanism of action and devices,"

Journal of Vascular and Interventional Radiology, vol. 21, no. 8, pp. S187-S191,

2010.

Page 51: Determining the Oncological and Immunological Effects of

14

[24] R. V. Davalos, L. Mir, and B. Rubinsky, "Tissue ablation with irreversible

electroporation," Annals of biomedical engineering, vol. 33, no. 2, p. 223, 2005.

[25] R. V. Davalos, B. Rubinsky, and D. M. Otten, "A feasibility study for electrical

impedance tomography as a means to monitor tissue electroporation for

molecular medicine," IEEE Transactions on Biomedical Engineering, vol. 49, no.

4, pp. 400-403, 2002.

[26] R. C. Martin et al., "Treatment of 200 locally advanced (stage III) pancreatic

adenocarcinoma patients with irreversible electroporation: safety and efficacy,"

Annals of surgery, vol. 262, no. 3, pp. 486-494, 2015.

[27] H. J. Scheffer et al., "Ablation of locally advanced pancreatic cancer with

percutaneous irreversible electroporation: results of the phase I/II PANFIRE

study," Radiology, vol. 282, no. 2, pp. 585-597, 2017.

[28] M. P. Belfiore et al., "Percutaneous CT-guided irreversible electroporation

followed by chemotherapy as a novel neoadjuvant protocol in locally advanced

pancreatic cancer: our preliminary experience," International Journal of Surgery,

vol. 21, pp. S34-S39, 2015.

[29] H. J. Scheffer et al., "Irreversible electroporation of locally advanced pancreatic

cancer transiently alleviates immune suppression and creates a window for

antitumor T cell activation," Oncoimmunology, vol. 8, no. 11, p. 1652532, 2019.

[30] R. C. Martin et al., "Irreversible electroporation in locally advanced pancreatic

cancer: a call for standardization of energy delivery," Journal of Surgical

Oncology, vol. 114, no. 7, pp. 865-871, 2016.

Page 52: Determining the Oncological and Immunological Effects of

15

[31] N. Beitel-White, R. C. G. Martin, Y. Li, R. M. Brock, I. C. Allen, and R. V.

Davalos, "Real-time prediction of patient immune cell modulation during

irreversible electroporation therapy," (in eng), Sci Rep, vol. 9, no. 1, p. 17739,

Nov 28 2019, doi: 10.1038/s41598-019-53974-w.

[32] R. M. Brock, N. Beitel-White, R. V. Davalos, and I. C. Allen, "Starting a Fire

Without Flame: The Induction of Cell Death and Inflammation in Electroporation-

Based Tumor Ablation Strategies," Frontiers in Oncology, vol. 10, 2020.

[33] Q. Shao et al., "Engineering T cell response to cancer antigens by choice of focal

therapeutic conditions," International Journal of Hyperthermia, vol. 36, no. 1, pp.

130-138, 2019.

[34] K. B. Bader, E. Vlaisavljevich, and A. D. Maxwell, "For whom the bubble grows:

physical principles of bubble nucleation and dynamics in histotripsy ultrasound

therapy," Ultrasound in medicine & biology, 2019.

[35] A. R. Smolock et al., "Robotically Assisted Sonic Therapy as a Noninvasive

Nonthermal Ablation Modality: Proof of Concept in a Porcine Liver Model,"

Radiology, vol. 287, no. 2, pp. 485-493, May 2018, doi:

10.1148/radiol.2018171544.

[36] E. Vlaisavljevich et al., "Non-invasive ultrasound liver ablation using histotripsy:

Chronic study in an in vivo rodent model," Ultrasound in medicine & biology, vol.

42, no. 8, pp. 1890-1902, 2016.

[37] E. Vlaisavljevich et al., "Non-invasive liver ablation using histotripsy: preclinical

safety study in an in vivo porcine model," Ultrasound in medicine & biology, vol.

43, no. 6, pp. 1237-1251, 2017.

Page 53: Determining the Oncological and Immunological Effects of

16

[38] E. Vlaisavljevich, Y. Kim, G. Owens, W. Roberts, C. Cain, and Z. Xu, "Effects of

tissue mechanical properties on susceptibility to histotripsy-induced tissue

damage," Physics in Medicine & Biology, vol. 59, no. 2, p. 253, 2013.

[39] E. Vlaisavljevich, Z. Xu, A. Arvidson, L. Jin, W. Roberts, and C. Cain, "Effects of

thermal preconditioning on tissue susceptibility to histotripsy," Ultrasound in

medicine & biology, vol. 41, no. 11, pp. 2938-2954, 2015.

[40] T. L. Hall, C. R. Hempel, K. Wojno, Z. Xu, C. A. Cain, and W. W. Roberts,

"Histotripsy of the prostate: dose effects in a chronic canine model," Urology, vol.

74, no. 4, pp. 932-937, 2009.

[41] A. Lake, Z. Xu, J. Wilkinson, C. Cain, and W. Roberts, "Renal ablation by

histotripsy—does it spare the collecting system?," The Journal of urology, vol.

179, no. 3, pp. 1150-1154, 2008.

[42] C. R. Hempel, T. L. Hall, C. A. Cain, J. B. Fowlkes, Z. Xu, and W. W. Roberts,

"Histotripsy fractionation of prostate tissue: local effects and systemic response

in a canine model," The Journal of urology, vol. 185, no. 4, pp. 1484-1489, 2011.

[43] E. Vlaisavljevich, Y. Kim, G. Owens, W. Roberts, C. Cain, and Z. Xu, "Effects of

tissue mechanical properties on susceptibility to histotripsy-induced tissue

damage," (in eng), Phys Med Biol, vol. 59, no. 2, pp. 253-70, Jan 20 2014, doi:

10.1088/0031-9155/59/2/253.

[44] E. Vlaisavljevich, A. Maxwell, M. Warnez, E. Johnsen, C. Cain, and Z. Xu,

"Histotripsy-induced cavitation cloud initiation thresholds in tissues of different

mechanical properties," (in eng), IEEE Transactions on Ultrasonics,

Page 54: Determining the Oncological and Immunological Effects of

17

Ferroelectrics, and Frequency Control, vol. 61, no. 2, pp. 341-52, Feb 2014, doi:

10.1109/TUFFC.2014.6722618.

[45] V. A. Khokhlova et al., "Histotripsy methods in mechanical disintegration of

tissue: Towards clinical applications," International journal of hyperthermia, vol.

31, no. 2, pp. 145-162, 2015.

[46] L. Thanos, S. Mylona, P. Galani, M. Pomoni, A. Pomoni, and I. Koskinas,

"Overcoming the heat-sink phenomenon: successful radiofrequency thermal

ablation of liver tumors in contact with blood vessels," Diagnostic and

Interventional Radiology, vol. 14, no. 1, p. 51, 2008.

[47] K. Pillai et al., "Heat sink effect on tumor ablation characteristics as observed in

monopolar radiofrequency, bipolar radiofrequency, and microwave, using ex vivo

calf liver model," Medicine, vol. 94, no. 9, 2015.

[48] C. M. Smolock AR, Vlaisavljevich E, Gendron-Fitzpatrick A, Green C, Ziemlewicz

TJ and Lee FT, "Robotically Assisted Sonic Therapy as a Non-Invasive Non-

Thermal Ablation Modality: Proof of Concept in a Porcine Liver Model,"

Radiology, vol. 287, no. 2, pp. 485-93, 2018.

[49] J. E. Coad, K. Kosari, A. Humar, and T. D. Sielaff, "Radiofrequency ablation

causes ‘thermal fixation’of hepatocellular carcinoma: a post‐liver transplant

histopathologic study," Clinical transplantation, vol. 17, no. 4, pp. 377-384, 2003.

[50] G. R. Schade, "Evaluation of the Systemic Response to Boiling Histotripsy

Treatment for Renal Carcinoma," 2018.

Page 55: Determining the Oncological and Immunological Effects of

18

[51] T. Worlikar et al., "Non-Invasive Orthotopic Liver Tumor Ablation Using

Histotripsy in an in Vivo Rodent Hepatocellular Carcinoma (HCC) Model,"

presented at the Society of Interventional Oncology, Boston, MA, USA, 2019.

[52] S. Qu et al., "Non-thermal histotripsy tumor ablation promotes abscopal immune

responses that enhance cancer immunotherapy," Journal for ImmunoTherapy of

Cancer, vol. 8, no. 1, 2020.

[53] A. Eranki et al., "High-Intensity Focused Ultrasound (HIFU) Triggers Immune

Sensitization of Refractory Murine Neuroblastoma to Checkpoint Inhibitor

Therapy," Clinical Cancer Research, vol. 26, no. 5, pp. 1152-1161, 2020.

[54] T. Worlikar et al., "Histotripsy for Non-Invasive Ablation of Hepatocellular

Carcinoma (HCC) Tumor in a Subcutaneous Xenograft Murine Model," Conf Proc

IEEE Eng Med Biol Soc, vol. 2018, pp. 6064-6067, Jul 2018, doi:

10.1109/EMBC.2018.8513650.

[55] G. R. Schade, Y.-N. Wang, S. D'Andrea, J. H. Hwang, W. C. Liles, and T. D.

Khokhlova, "Boiling Histotripsy Ablation of Renal Cell Carcinoma in the Eker Rat

Promotes a Systemic Inflammatory Response," Ultrasound in medicine &

biology, vol. 45, no. 1, pp. 137-147, 2019.

[56] T. Worlikar et al., "Non-invasive liver cancer ablation using histotripsy in an in

vivo murine hepatocellular carcinoma (HCC) model," in Ultrasonics Symposium

(IUS), 2017 IEEE International, 2017: IEEE, pp. 1-1.

[57] K. J. Pahk et al., "Boiling Histotripsy-induced Partial Mechanical Ablation

Modulates Tumour Microenvironment by Promoting Immunogenic Cell Death of

Cancers," Scientific reports, vol. 9, no. 1, pp. 1-12, 2019.

Page 56: Determining the Oncological and Immunological Effects of

19

[58] S. X. Vidal Jove J, Vlaisavljevich E, Cannata J, Duryea A, Miller R, Lee F and

Ziemlewicz T, "Phase I Study of Safety and Efficacy of Hepatic Histotripsy:

Preliminary Results of First in Man Experience with Robotically-Assisted Sonic

Therapy," in International Society of Therapeutic Ultrasound, Barcelona, Spain,

2019.

[59] V. E. Kim Y, Owens GE, Allen SP, Cain CA and Xu Z, "In vivo transcostal

histotripsy therapy without aberration correction," Phys Med Biol, vol. 59, pp.

2553-68, 2014.

[60] E. Vlaisavljevich et al., "Image-guided non-invasive ultrasound liver ablation

using histotripsy: feasibility study in an in vivo porcine model," Ultrasound Med

Biol, vol. 39, no. 8, pp. 1398-409, Aug 2013, doi:

10.1016/j.ultrasmedbio.2013.02.005.

[61] E. Vlaisavljevich et al., "Non-Invasive Liver Ablation Using Histotripsy: Preclinical

Safety Study in an In Vivo Porcine Model," Ultrasound Med Biol, vol. 43, no. 6,

pp. 1237-1251, Jun 2017, doi: 10.1016/j.ultrasmedbio.2017.01.016.

[62] T. Flisikowska, A. Kind, and A. Schnieke, "Pigs as models of human cancers,"

Theriogenology, vol. 86, no. 1, pp. 433-7, Jul 1 2016, doi:

10.1016/j.theriogenology.2016.04.058.

[63] K. M. Schachtschneider et al., "The Oncopig Cancer Model: An Innovative Large

Animal Translational Oncology Platform," Front Oncol, vol. 7, p. 190, 2017, doi:

10.3389/fonc.2017.00190.

[64] L. B. Schook et al., "A Genetic Porcine Model of Cancer," PLoS One, vol. 10, no.

7, p. e0128864, 2015, doi: 10.1371/journal.pone.0128864.

Page 57: Determining the Oncological and Immunological Effects of

20

[65] K. Lee, K. Farrell, and K. Uh, "Application of genome-editing systems to enhance

available pig resources for agriculture and biomedicine," (in eng), Reprod Fertil

Dev, vol. 32, no. 2, pp. 40-49, Jan 2019, doi: 10.1071/rd19273.

[66] J. J. Tentler et al., "Patient-derived tumour xenografts as models for oncology

drug development," (in eng), Nature reviews. Clinical oncology, vol. 9, no. 6, pp.

338-50, Apr 17 2012, doi: 10.1038/nrclinonc.2012.61.

[67] R. M. Brock et al., "Patient derived xenografts expand human primary pancreatic

tumor tissue availability for ex vivo irreversible electroporation testing," Frontiers

in Oncology, vol. 10, 2020.

[68] E. Vlaisavljevich et al., "Non-Invasive Ultrasound Liver Ablation Using Histotripsy:

Chronic Study in an In Vivo Rodent Model," Ultrasound Med Biol, vol. 42, no. 8,

pp. 1890-902, Aug 2016, doi: 10.1016/j.ultrasmedbio.2016.03.018.

[69] S. S. Pinho, S. Carvalho, J. Cabral, C. A. Reis, and F. Gartner, "Canine tumors: a

spontaneous animal model of human carcinogenesis," Transl Res, vol. 159, no.

3, pp. 165-72, Mar 2012, doi: 10.1016/j.trsl.2011.11.005.

[70] J. D. Schiffman and M. Breen, "Comparative oncology: what dogs and other

species can teach us about humans with cancer," Philos Trans R Soc Lond B

Biol Sci, vol. 370, no. 1673, Jul 19 2015, doi: 10.1098/rstb.2014.0231.

[71] M. Paoloni et al., "Defining the Pharmacodynamic Profile and Therapeutic Index

of NHS-IL12 Immunocytokine in Dogs with Malignant Melanoma," PLoS One, vol.

10, no. 6, p. e0129954, 2015, doi: 10.1371/journal.pone.0129954.

[72] M. Paoloni et al., "Prospective molecular profiling of canine cancers provides a

clinically relevant comparative model for evaluating personalized medicine

Page 58: Determining the Oncological and Immunological Effects of

21

(PMed) trials," PLoS One, vol. 9, no. 3, p. e90028, 2014, doi:

10.1371/journal.pone.0090028.

[73] M. Paoloni, S. Lana, D. Thamm, C. Mazcko, and S. Withrow, "The creation of the

Comparative Oncology Trials Consortium Pharmacodynamic Core: Infrastructure

for a virtual laboratory," Vet J, vol. 185, no. 1, pp. 88-9, Jul 2010, doi:

10.1016/j.tvjl.2010.04.019.

[74] M. C. Paoloni et al., "Rapamycin pharmacokinetic and pharmacodynamic

relationships in osteosarcoma: a comparative oncology study in dogs," PLoS

One, vol. 5, no. 6, p. e11013, Jun 08 2010, doi: 10.1371/journal.pone.0011013.

[75] I. Gordon, M. Paoloni, C. Mazcko, and C. Khanna, "The Comparative Oncology

Trials Consortium: using spontaneously occurring cancers in dogs to inform the

cancer drug development pathway," PLoS Med, vol. 6, no. 10, p. e1000161, Oct

2009, doi: 10.1371/journal.pmed.1000161.

[76] M. C. Paoloni et al., "Launching a novel preclinical infrastructure: comparative

oncology trials consortium directed therapeutic targeting of TNFalpha to cancer

vasculature," PLoS One, vol. 4, no. 3, p. e4972, 2009, doi:

10.1371/journal.pone.0004972.

[77] C. A. London et al., "Preclinical evaluation of the novel, orally bioavailable

Selective Inhibitor of Nuclear Export (SINE) KPT-335 in spontaneous canine

cancer: results of a phase I study," PLoS One, vol. 9, no. 2, p. e87585, 2014, doi:

10.1371/journal.pone.0087585.

Page 59: Determining the Oncological and Immunological Effects of

22

Chapter 2. Histotripsy for the Treatment of Cholangiocarcinoma Liver Tumors: In Vivo Feasibility and Ex Vivo Dosimetry Study

Alissa Hendricks, Peter Weber, Alex Simon, Sofie Saunier, Sheryl Coutermarsh-Ott, Douglas Grider, Joan Vidal Jove, Irving Coy Allen, David Luyimbazi, Eli Vlaisavljevich

This chapter is excerpted from a manuscript published in IEEE Transactions in UFFC.

2021 IEEE. Reprinted, with permission, from: A. Hendricks-Wenger et al., "Histotripsy for the Treatment of Cholangiocarcinoma Liver Tumors: In Vivo Feasibility and Ex Vivo Dosimetry Study," in IEEE Transactions on Ultrasonics, Ferroelectrics, and Frequency

Control, doi: 10.1109/TUFFC.2021.3073563.

Page 60: Determining the Oncological and Immunological Effects of

23

2.1 Abstract

Histotripsy is a non-invasive, non-ionizing, and non-thermal focused ultrasound

ablation method that is currently being developed for the treatment of liver cancer.

Promisingly, histotripsy has been shown for ablating primary (hepatocellular carcinoma,

HCC) and metastatic (colorectal liver metastasis, CLM) liver tumors in preclinical and

early clinical studies. The feasibility of treating cholangiocarcinoma (CC), a less

common primary liver tumor that arises from the bile ducts, has not been explored

previously. Given that prior work has established that histotripsy susceptibility is based

on tissue mechanical properties, there is a need to explore histotripsy as a treatment for

CC due to their dense fibrotic stromal components. In this work, we first investigated the

feasibility of histotripsy for ablating CC tumors in vivo in a patient-derived xenograft

mouse model. The results showed that histotripsy could generate CC tumor ablation

using a 1 MHz small animal histotripsy system with treatment doses of 250, 500, and

1000 pulses/point. A second set of experiments compared the histotripsy doses

required to ablate CC tumors to HCC and CLM tumors ex vivo. For this, human tumor

samples were harvested after surgery and treated ex vivo with a 700 kHz clinical

histotripsy transducer. Results demonstrated significantly higher treatment doses were

required to ablate CC and CLM tumors compared to HCC, with the highest treatment

dose required for CC tumors. Overall, the results of this study suggest that histotripsy

has the potential to be used for the ablation of CC tumors while also highlighting the

need for tumor-specific treatment strategies.

Page 61: Determining the Oncological and Immunological Effects of

24

2.2 Keywords:

Focused Ultrasound, Histotripsy, Liver Cancer, Cholangiocarcinoma, Hepatocellular

Carcinoma, Liver Metastasis

Page 62: Determining the Oncological and Immunological Effects of

25

2.3 Introduction

More than 800,000 people are diagnosed with a malignancy originating from or

metastatic to the liver every year. Of these, 42,810 cases are projected for the U.S. in

2020 [1]. These malignant liver tumors include hepatocellular carcinoma (HCC), liver

metastasis (LM), and cholangiocarcinoma (CC). Although surgical resection remains the

mainstay of liver tumor intervention, less than 25% of patients are candidates for

resection due to tumor burden, location, or comorbid disease [2, 3]. For example, a prior

study found that only 15% of intrahepatic CC tumors and 56% of distal tumors were

deemed resectable [4].

Minimally-invasive ablation methods have shown some success in treating liver

tumors but have limitations due to the thermal mechanism, including lack of precision

and high complication rates when treating near critical structures [5-14]. Histotripsy is a

non-invasive, non-thermal, and image-guided focused ultrasound ablation method that

ablates tissue through the generation of acoustic cavitation at the transducer focus [15-

19]. Histotripsy uses microsecond long, high-pressure pulses to generate a histotripsy

bubble cloud inside of the targeted tissue [20-24]. The rapid expansion and collapse of

the cavitation bubbles results in complete disruption of the tissue [25]. As a non-thermal

ablation modality, histotripsy has shown the potential to overcome the limitations

associated with thermal ablation. For instance, hepatic histotripsy has been shown to

produce consistent and complete ablation in highly vascular regions of the liver [18, 26].

Histotripsy has also shown the ability to preserve vital structures such as major vessels,

Page 63: Determining the Oncological and Immunological Effects of

26

bile ducts, and nerves within the ablation zone [18, 27]. Preclinical in vivo studies of

hepatic histotripsy in healthy porcine and rodent models have demonstrated that

histotripsy can generate clinically relevant ablation zones with sharp boundaries (<1mm)

between treated and healthy liver tissue [15, 17, 18, 26, 27]. Additional studies have

shown the in vivo feasibility of treating HCC tumors in rodent models using histotripsy

[28, 29]. Based on these successful preclinical studies, a Phase I clinical trial in patients

with liver cancer was recently conducted to demonstrate the feasibility of hepatic

histotripsy [16]. Together, these studies have established the feasibility and overall

safety profile of hepatic histotripsy.

Although one patient with CC was reportedly treated as part of the clinical trial

mentioned above, there have been limited studies investigating the potential of

histotripsy for treating CC tumors. CC tumors pose a unique challenge for ablation

modalities due to their common location near vital critical structures [30]. The vessel

and duct sparing features of hepatic histotripsy are promising for the treatment of CC

located near these structures. However, given prior studies showing fibrous tissues with

higher stiffness and mechanical strength are more resistant to histotripsy, it is also

expected that CC tumors may pose a unique challenge for histotripsy. This hypothesis

is based on the CC tumor’s dense desmoplastic fibrotic composition and higher stiffness

that has been reported previously [31-33]. For instance, prior work has found the

average Young’s modulus of CC tumors to be 56.9±25.6 kPa, which was significantly

higher than HCC (14.86±10.0 kPa) and LM (28.8±16.0 kPa) tumors [34]. Given these

mechanical profiles, we hypothesize that histotripsy will require higher treatment doses

to ablate CC tumors compared to LM and HCC.

Page 64: Determining the Oncological and Immunological Effects of

27

In this study, we investigate the feasibility of using histotripsy for the treatment of

CC tumors. To test histotripsy for CC ablation, this study was split into two parts. In part

one, we investigate the in vivo feasibility of histotripsy CC ablation in a subcutaneous

patient-derived xenograft (PDX) CC tumor model. In part two, we perform a comparative

dosimetry study for treating ex vivo human HCC, LM, and CC tumors with histotripsy to

determine the differences in treatment dose requirements for ablating each tumor type

with histotripsy.

2.4 Procedures

2.4.1 Histotripsy Systems

Two separate histotripsy systems were used in this study for the small animal and

large animal in vivo histotripsy studies. Small animal in vivo experiments were

conducted using a custom 8-element 1MHz histotripsy transducer with a geometric focal

distance of 36 mm, an aperture size of 52.7 mm, and an f-number of 0.68 (Fig.1B). The

transducer was driven via a custom high-voltage pulser designed to generate short

therapy pulses of <2 cycles controlled by a field-programmable gate array (FPGA)

board (Altera DE0-Nano Terasic Technology, Dover, DE, USA) programmed for

histotripsy therapy pulsing. The transducer was positioned in a tank of degassed water

beneath a custom-designed mouse surgical stage (Fig.1) and attached to a computer-

guided 3-D positioning system with a 0.05 mm motor resolution to control the automated

volumetric treatments. A linear ultrasound imaging probe with a frequency range of 10-

Page 65: Determining the Oncological and Immunological Effects of

28

18 MHz (L18-10L30H-4, Telemed, Lithuania, EU) was coaxially aligned inside the

transducer for treatment guidance and monitoring [28, 29]. The transducer was powered

by a high voltage DC power supply (GENH750W, TDK-Lambda), and the system was

controlled using a custom user-interface operated through MATLAB (MathWorks).

For the ex vivo experiments, histotripsy was applied using a 36-element 700 kHz

clinical histotripsy transducer and associated driving systems (HistoSonics, Inc) that

was previously used in a human clinical trial for the treatment of benign prostatic

hyperplasia [35] and multiple previous pre-clinical studies [36, 37]. This system is the

same frequency as the clinical histotripsy system used in the recent Phase I human liver

cancer trial [16] and was thus chosen for this study. The transducer that consists of a

concave circular aperture with a small inlet on the perimeter to allow for histotripsy

treatments to be guided by a trans-rectal imaging probe (Note: this imaging probe was

not used in the current study [36, 37]. The transducer has a geometric focal distance of

11 cm and FWHM dimensions at a geometric focus of 4.0 mm, 4.0 mm, and 15.0 mm in

the transverse, elevational, and axial dimensions, respectively. The transducer was

powered by a high voltage DC power supply (GENH750W, TDK-Lambda), and the

system was controlled using a custom user-interface called the Histotripsy Service Tool

(HistoSonics, Inc.) to produce 5-cycle histotripsy pulses [24, 38].

2.4.2 Pressure Calibration

Focal pressure waveforms for the 1 MHz and 700 kHz transducers were

measured by a high sensitivity rod hydrophone (HNR-0500, Onda Corporation,

Page 66: Determining the Oncological and Immunological Effects of

29

Sunnyvale, CA, USA) and a custom-built fiber optic probe hydrophone (FOPH) [39, 40]

in degassed water at the focal point of each transducer. Focal pressures were directly

measured with the FOPH up to a peak negative pressure (p-) of ~16MPa for the 700kHz

transducer and ~20 MPa for the 1MHz transducer. Pressures were not directly

measured beyond these limits due to cavitation on the tip of the hydrophone probe.

Waveforms at higher p- were estimated using linear summation of the pressure

waveforms directly measured from four sub-apertures of the array for the 700 kHz

transducer, and two sub-apertures for the 1MHz transducer. This method showed close

agreement between directly measured pressures and the cavitation threshold seen

experimentally [41].All waveforms were measured using a Tektronix TBS2000 series

oscilloscope at a sample rate of 500MS/s, with the waveforms averaged over 128

pulses and recorded in MATLAB.

2.4.3 Histotripsy In Vivo CC Ablation Procedure

The feasibility of histotripsy for CC tumor ablation was tested in vivo using a

patient-derived xenograft (PDX) CC mouse model. Twelve female NSG mice with

subcutaneous flank PDX CC tumors were ordered from and prepared by Jackson

Laboratory (Bar Harbor, Maine, USA). Through a process previously described for

pancreatic PDX tumors [42], a human CC tumor was collected from a patient and

propagated as a heterogeneous tumor in immunocompromised NSG mice. Once

received, animals were monitored three times weekly under Virginia Tech IACUC

protocols. At each check, general health was monitored and tumor diameter was

Page 67: Determining the Oncological and Immunological Effects of

30

determined by taking two perpendicular measurements, one of which was the widest

point of the tumor. A single tumor diameter was calculated as the square root of the

product of the two measurements. The mice underwent histotripsy treatment when their

tumors reach the targeted treatment size of ~5mm in diameter. Before treatment, fur

was removed over the tumor and the surrounding area by applying Nair (Naircare,

Ewing, NJ, USA) for 1-2 minutes and removed by wiping off with a wet paper towel. For

treatment, mice were anesthetized with isoflurane (1.5 L/min oxygen flow rate with 1.0-

2.5% isoflurane) and placed prone on the subject stage (Fig.1) with their tumor

submerged underwater. Animals’ respiratory rate and rhythm were monitored during

Figure 2-1 In Vivo Ablation Setup. (A) Histotripsy murine experiments were conducted using a custom 1MHz small animal histotripsy system. (B) Subject’s tumors were submerged in degassed water at the transducer focus in order for (C) histotripsy to be applied to the tumor non-invasively guided by real-time ultrasound imaging.

Page 68: Determining the Oncological and Immunological Effects of

31

treatment by trained personnel.

The overall setup for the in vivo treatment is illustrated in Figure 1. This custom

system, which has been used in previous small animal in vivo histotripsy studies [17,

28, 29], consists of the 1 MHz histotripsy transducer with co-axially aligned ultrasound

imaging positioned vertically underneath a small animal surgical stage inside a heated

water tank that allows for acoustic coupling. A second water tank is connected to this

primary tank to circulate warmed, degassed water at a constant temperature throughout

the procedures. Before treatment, the water tanks were filled with water and degassed

for approximately 1 hour. A water heater was used to maintain the water temperature at

37+4oC throughout treatment. A small animal platform containing a hole in the center

was located on top of the tank for an anesthetized mouse to be placed with the mouse’s

flank containing the tumor submerged in the water at the transducer focus. The

transducer and a co-axially aligned imaging probe were connected to a 3D motorized

positioning system that was used to apply an automated volumetric ablation to a

targeted volume within the tumor. This custom volumetric ablation system was designed

to allow the user to outline the desired tumor and treatment regions before starting

treatment. The treatment volume was then divided into a 3D grid of points, and

histotripsy was raster-scanned through a series of treatment points spaced by 1.5 mm

in the axial direction and 0.5 mm in the lateral directions. While all the tumors were

treated at different sizes due to delayed growth and planning in between treatments, the

goal was to treat tumors when they averaged 5 mm in diameter. For a 5 mm diameter

tumor, the spacing resulted in a treatment with 1382 treatment points, a dwell time of 2

seconds, and a 46-minute treatment. This treatment spacing was determined based on

Page 69: Determining the Oncological and Immunological Effects of

32

preliminary experiments to ensure overlap between adjacent bubble clouds. Three

separate treatment doses of 250, 500, and 1000 pulses/point were tested to determine

the feasibility of histotripsy for treating CC tumors in vivo, with the results compared to

an untreated group (n=3/group). Treatment dose was determined based on prior small

animal studies that utilized 50-500 pulses to treat healthy liver and HCC tumors in

addition to the hypothesis that CC tumors would require a high dose given its higher

mechanical strength [17, 28, 29].

2.4.4 Human Liver Tumor Specimens for Ex Vivo Treatments

Liver tumor specimens were obtained by consent from patients with resectable

HCC, CLM, or CC tumors in accordance with the Institutional IRB approved protocol

Figure 2-2 Ex Vivo Tissue Ablation Setup. Histotripsy dose experiments were conducted using a 700kHz transducer with the focus aligned in the center of the targeted tissue, with guidance from ultrasound imaging aligned orthogonally (A-B). Tissues were imbedded within the center of a gelatin phantom (C).

Page 70: Determining the Oncological and Immunological Effects of

33

(Carilion Clinic, Roanoke Memorial Hospital, Roanoke, VA). Patients with either biopsy-

proven or radiologically diagnosed liver tumors were consented and enrolled in the

study. At the time of resection, the resected tissues were sent fresh to pathology for

processing, and any tissue above what was needed for clinical evaluation was donated

to this study. When possible, surrounding liver parenchyma (LP) was also collected and

used to evaluate the histotripsy doses for ablating LP tissue within the surgical margin.

After harvesting, the tumor specimens were placed in a 0.9% degassed saline solution.

Before treatment, samples were degassed inside a saline solution in a vacuum chamber

(20-25 kPa) for 30-60 minutes. After degassing, the harvested tumor samples were

sectioned into 0.5-1cm3 samples and embedded in 7.5% degassed gelatin (porcine

gelatin, Millipore Sigma) tissue phantoms for treatment (Fig.2C). Gel phantoms

containing the embedded tumor samples were then refrigerated for up to 8 hours, with

all specimens treated within 24 hours of the initial harvest.

2.4.5 Histotripsy Ex Vivo Ablation Procedure

The treatment doses required to ablate HCC, CLM, and CC tumors were

investigated by histotripsy applying to the excised tissue samples embedded inside

gelatin phantoms, as described above (Fig.2). For all experiments, the 700 kHz

histotripsy therapy transducer was used to apply 5 cycle pulses to the center of the

tissue samples at a PRF of 500 Hz. These parameters closely match the histotripsy

parameters used in previous shock-scattering histotripsy liver ablation studies [15, 16,

18, 27]. For each treatment, tissue samples embedded in gelatin were mounted to a

Page 71: Determining the Oncological and Immunological Effects of

34

motorized 3D positioning system and aligned to the focus of the therapy transducer

(Fig.2). To guide treatments and visualize the histotripsy cavitation bubble cloud in real-

time, an ultrasound imaging probe (SL18-5, AIXPLORER, SuperSonic Imagine) was

mounted perpendicularly to the tissue and aligned to the focal location of the therapy

transducer (Fig.2B). The focus of the histotripsy transducer was located and marked on

the imaging screen before each experiment by generating a bubble cloud in open water

and then marking the location on the imaging screen before aligning the tissue at the

focus. Tissue samples were then positioned such that the focal point of the transducer

was located at the desired location within the tissue.

The pressure required to generate histotripsy cavitation inside each tissue

sample was measured by gradually increasing the pressure at the focus until a

cavitation cloud was identified on US imaging. The pressure at which a consistent

histotripsy bubble cloud was generated inside each tissue sample was recorded as the

cavitation cloud threshold before conducting volumetric ablation experiments in each

sample.

Histotripsy was applied to the targeted volume at a pressure level chosen to be

~30% above the cavitation threshold, which corresponded to treatments with applied p-

between 19 MPa and 33 MPa, depending upon the sample. Volumetric treatments were

then applied automatically to a predetermined 3D grid of equally spaced treatment

points, with 2.5 mm spacing in the axial direction and 1 mm spacing in the later

directions to ensure overlap between adjacent bubble clouds. For each experiment, this

grid of points was treated with histotripsy by mechanically scanning the therapy focal

zone to cover the targeted portion of the tumor sample. The ideal treatment was a 5mm

Page 72: Determining the Oncological and Immunological Effects of

35

x 5mm x 5mm cube consisting of 75 total points at which to dwell. Given that some

samples were too small to achieve this with a contained ablation volume, not all

ablations were this large. The desired size for each sample was 15mm x 15mm x

15mm. However, due to the irregular shapes of the tumors and the small size of some

samples, this was not always achievable. The smallest sample treated in this study was

a CC tumor that was approximately 8mm x 8mm x 8mm, which led to a treatment size

of 2.5mm x 3mm x 3mm.

For each tumor type, three to five groups were treated with histotripsy at different

doses to characterize the extent of tissue ablation generated at different histotripsy

doses (i.e., number of pulses/point). The number of pulses applied per point ranged

from 500-2000 pulses/point for HCC and CLM tumors and 1000-4000 pulses/point for

CC tumors. These treatment doses were chosen based on previous liver histotripsy

studies [15, 36, 43], with the higher doses used for CC tumors based upon a

combination of previous histotripsy studies on excised tissues [44], the relationship

between histotripsy susceptibility and mechanical properties [31, 32, 44], and literature

showing CC have an increased Young’s modulus [34]. In addition to the liver tumor

ablation experiments, a separate set of treatments was conducted on LP tissues

collected from the surgical margins around the tumors at doses of 250-1500

pulses/point. A sample size of n=3 per tissue type and treatment dose was used for all

Page 73: Determining the Oncological and Immunological Effects of

36

experiments when possible. However, due to limitations in tissue availability, this

sample size could not be achieved for all tissues and all doses. Based on the availability

of donated tissue samples, a total of 49 experimental treatments were conducted in this

study including 8 HCC tumor samples, 15 CLM tumor samples, 12 CC tumor samples,

and 9 LP tissue samples. For each tissue, the sample numbers treated per dose is

listed in Table 1.

Table 2-1 Ex Vivo sample numbers by tissue type and Dose

2.4.6 Histology & Morphological Analysis

After treatment, tissue specimens were visually inspected and fixed in 10% formalin

for at least 24 hours before sectioning and staining. Given that each treatment was

focused on the center portion of the tissue, the central region of each tissue was

sectioned into a cassette. Using standard protocols, all tissues were paraffin embedded

and stained using hematoxylin and eosin (H&E), by ViTALS (Virginia Tech Animal

Laboratory Services, Blacksburg, VA), to assess for histotripsy damage to the tissue

parenchyma and any other changes in structure and density of collagen and other

Pulses/Point Dose

250 500 1000 1500 2000 4000

HCC n=2 n=2 n=1

CLM n=3 n=3 n=3

CC n=3 n=3 n=3 n=2

LP n=3 n=4 n=2

Page 74: Determining the Oncological and Immunological Effects of

37

tissue structures within the ablation volume. To determine the estimated extent of tissue

ablated within the targeted volume, regions of cellular damage were compared with

regions that were outside of the ablation zone. Results were then verified by a

veterinary pathologist (SCO) and a human pathologist (DG) to ensure accuracy and

avoid biases. Histology images are representative of the samples treated.

2.5 Results

2.5.1 Histotripsy Ablation of Subcutaneous PDX CC Tumors

Well–defined histotripsy bubble clouds were observed at the focal locations in all

tumors at the beginning of the treatment (Fig.1B). Throughout the automated volumetric

treatments, the bubble clouds were continuously observed at the focus as dynamically

changing hyperechoic regions within the tissue. After treatment, histotripsy ablation

zones could be visualized on ultrasound imaging as hypoechoic regions that

corresponded with the regions exposed to the bubble cloud. This was true at each of

the three doses tested, with more clearly defined hypoechoic regions observed on

ultrasound imaging as the treatment dose increased from 250 to 1000 pulses/point

(Fig.3B-D). Histological analysis of the ablation zones showed complete ablation (no

remaining intact cells) of the CC tumors within the center of the treated region after 250

pulses/point treatments (Fig.3J). Similarly, results for the 500 pulses/point and 1000

pulses/point treatment groups showed complete ablation of the CC tumors inside the

treated regions (Fig3.K-L). The histological analysis also showed more well-defined

Page 75: Determining the Oncological and Immunological Effects of

38

treatment margins with increasing treatment dose (Fig.3F-H). More specifically, results

from the 250 pulses/point and 500 pulses/point treatments showed small (<150µm)

regions of disrupted tissue on the margins between ablated and intact tissue that

included intact cells within the boundary region (Fig.3F-G). In contrast, these partially

treated boundary regions were not observed for the 1000 pulses/point treatments that

contained a well-defined boundary between the completely treated region and the

surrounding intact tissue (Fig.3H).

Figure 2-3 Dotted orange circles on US images (A-D) indicate approximate region of ablation. Low magnification H&E images shows ablation margins (E-H). High magnification H&E images of control and ablated region (J-L). Black boxes on 10x images indicate area magnified in respective 40x images. Scale bar on 10x H&E images = 100um (E-H), 40x scale bar = 50um (I-J).

Page 76: Determining the Oncological and Immunological Effects of

39

2.5.2 Histotripsy Bubble Cloud Generation in Ex Vivo Tissue Specimens

The p- required to generate histotripsy bubble clouds inside each of the different

tissue types using the 700kHz clinical histotripsy transducer was measured to be

22.2±1.2 MPa, 17.7±3.1 MPa, 16.5±2.6 MPa, and 20.6+4.6 for HCC, CLM, CC, and LP

samples, respectively. In general, the bubble clouds in all tissue types showed typical

histotripsy features with a dense dynamically changing hyperechoic bubble cloud

observed within or near the focal region of the transducer, as visualized on US imaging.

During volumetric ablation experiments, the hyperechoic bubble clouds were maintained

throughout the treatments in all tissue experiments. In some samples, particularly

samples smaller than the desired 15mm x 15 mm x 15 mm dimensions, the cavitation

bubble clouds formed at the interface of the gelatin phantom and the tissue including

instances of both prefocal and post-focal cavitation clouds. These effects were

observed more often in the CLM and CC samples. In cases in which prefocal cavitation

was noted during the initial bubble cloud generation, the treatment zone was adjusted to

a deeper location in the sample when possible and the volumetric treatments were only

included as part of this study for samples in which a focal bubble cloud remained

present for the duration of the treatment.

2.5.3 Histotripsy Ablation of Ex Vivo HCC Specimens

For HCC samples treated with 500 or more pulses/point (Fig.4B-D/F-H), histotripsy

resulted in complete ablation of the targeted tumor volume with no notable regions of

intact cells remaining inside the ablated area. There was tumor necrosis characterized

Page 77: Determining the Oncological and Immunological Effects of

40

by remaining cell nuclei and other subcellular tissue debris inside the treated regions on

these samples, with the number of these nuclei reduced in concentration for the higher

treatment doses. At 1000 pulses/point or more, there was a complete loss of cellular

structures characterized as tissue ablated to amorphous debris.

2.5.4 Histotripsy Ablation of Ex Vivo CLM Specimens

Histological analysis demonstrated partial tumor ablation for CLM samples treated

with 500 pulses/point, with more than 75% of cells within the treated region remaining

after treatment (Fig.5B/G). Small, sporadic foci of ablation were observed inside of

these samples, with the majority of the treatment volume remaining intact. For 1000

pulses/point CLM samples, a larger portion of the tumor samples were observed to be

ablated after treatment with ~25-75% of the targeted region containing viable tumor

Figure 2-4 Comparison of untreated human HCC tumors with ex vivo ablations at 500 pulses/point, 1000 pulses/point, and 2000 pulses/point. 10x magnification scale bar= 100um (A-D), 40x scale bar= 50um (E-H). Black boxes on 10x images indicate area magnified in respective 40x images. Histology images are representative of the samples treated.

Page 78: Determining the Oncological and Immunological Effects of

41

tissue after histotripsy (Fig.5C/H). At the higher treatment doses of 1500 and 2000

pulses/point, histotripsy was observed to generate significantly more tissue damage

with complete ablation of the targeted tumor regions observed in some of the samples

at both doses (Fig.5D-E/I-J). However, results showed a large amount of variability in

the extent of ablation among treatments within each group, with some samples showing

only partial tumor ablation. More specifically, the number of viable cells remaining after

treatment ranged from <5% (complete ablation) to ~25% in each group, with complete

ablation achieved in 2 of the 3 samples at 1500 pulses/point and 1 of the 2 samples

tested at 2000 pulses/point. It was noted that the tissues that did not show complete

Figure 2-5 Comparison of untreated human CLM tumors with those treated at 500 pulses/point, 1000 pulses/point, 1500 pulses/point, and 2000 pulses/point. 10x magnification scale bar= 100um (A-E), 40x scale bar= 50um (F-J). Black boxes on 10x images indicate area magnified in respective 40x image. Histology images are representative of the samples treated.

Page 79: Determining the Oncological and Immunological Effects of

42

ablation within these groups appeared to contain more glandular structures, thus, were

more well-differentiated. Less differentiated tumors with poor cellular organization had

greater ablation. Similarly, histological analysis showed the CLM tumors tested in this

study exhibited varying degrees of fibrosis within the tumor. Partial ablations were most

often exhibited by CLM tumors with a robust, desmoplastic (or fibrous) response. For

example, for the 1000 pulses/point samples shown in Figure 5, the ablated regions of

the tumor are found sporadically within the more fibrous regions of the tissue showing

loss of cellular detail but still maintaining the fibrotic tissue architecture (Fig.5C/H). In

contrast, the extent of ablation was significantly higher within the less fibrotic regions of

these tumors. This effect can be seen in the example 2000 pulses/point image shown in

Figure 5, in which large regions of the tissue were fully ablated, with the remaining

Figure 2-6 Comparison of untreated human CC tumors with those treated at 1000 pulses/point, 1500 pulses/point, 2000 pulses/point, and 4000 pulses/point. 10x magnification scale bar= 100um (A-E), 40x scale bar= 50um (F-J). Black boxes on 10x images indicate area magnified in respective 40x image. Histology images are representative of the samples treated.

Page 80: Determining the Oncological and Immunological Effects of

43

tissue lacking cellular detail but with a few nuclei corresponding to the more fibrotic

portions of the tumor.

2.5.5 Histotripsy Ablation of Ex Vivo CC Specimens

Histological analysis demonstrated partial tumor ablation for CC samples treated

with 1000 pulses/point, with sporadic foci of ablated tumor observed throughout the

treated volume with interspersed regions of intact tumor (Fig.6B/G). Results for the

1500 pulses/point and 2000 pulses/point treatments showed similar results with larger

regions of complete loss of cellular detail, supporting a completely ablated portion of the

tumor in these samples (Fig.6) compared to the 1000 pulses/point group (Fig.6B/G).

For example, the 2000 pulses/point sample shown in Figure 6 has two visible regions of

tumor that were not fully ablated, both of which correspond to more fibrotic regions of

the tissue. In contrast, the fully ablated regions within this sample appear to correspond

to less dense and less fibrotic regions of the tumor. At the highest treatment dose of

4000 pulses/point, histotripsy again resulted in a similar result with only partial ablation

generating within each of the CC tumors and ~50-75% of the cells showing viable nuclei

but with loss of cytoplasmic detail within the ablation region remaining after treatment

(Fig.6E/J). In these samples, the sporadic foci of ablation again appeared to be located

in the regions of the tissue low in fibrosis (Fig.6). Looking at these foci under higher

magnification, the tissue within these ablated regions showed complete ablation with no

remaining viable cells. In contrast, the less ablated regions of the CC tumors were

highly more fibrotic with >75-90% of viable cells remaining after treatment, as shown in

Page 81: Determining the Oncological and Immunological Effects of

44

Supplemental Figure S1. Finally, it is important to note the high amount of variability in

the extent of ablation was observed among treatments within each group. Several of the

treatments resulted in >75% ablation (<25% of viable cells remaining) at dosing

regimens of 1000, 1500, 2000, and 4000 pulses/point whereas other treatments

resulted in <25% ablation with the majority of the targeted region remaining intact after

treatment.

2.5.6 Histotripsy Ablation of Ex Vivo LP Specimens

Part of the surrounding liver parenchyma was excised in addition to the tumor for six

patients enrolled in this study. Four patients had hepatic steatosis as identified by

abdominal imaging. One patient had hemochromatosis in addition to steatosis, one was

Hepatitis C positive with fibrosis, and another patient had no identifiable liver disease.

These samples were divided based on the presence of steatosis. On histology, tissues

Figure 2-7 Supplemental Figure S1. Single sample of Cholangiocarcinoma Ablated at 4000 pulses/point illustrating fibrosis dependence to susceptibility. Region of fibrosis (A) showed no damage, while less fibrotic region (B) had sporadic ablated foci. High magnification of the region in B indicated by the black box is Figure 7E 4000 pulses/point location. Scale bars = 500um. * Indicate ablated region.

Page 82: Determining the Oncological and Immunological Effects of

45

from patients with hepatic steatosis were readily apparent based on the presence of

macrovesicular fat globules within the tissues. For non-steatotic samples, a sample size

of n=1 was conducted at 250, 500, and 1000 pulses/point. Steatotic samples were

treated at n=2 at 250 pulses/point, n=3 for 500 pulses/point, and n=1 for 1000

pulses/point. A complete ablation was formed within the tissues that did not have

hepatic steatosis even at doses as low as 250 pulses/point (Fig.7). In comparison,

tissues with hepatic steatosis were noted to have sporadic foci of ablations at 250

pulses/point with more complete ablation generated at the higher doses. For instance,

only a few remaining viable cells were visible inside the ablation zone after 500

pulses/point and complete ablation of the treated region was observed after 1000

pulses/point (Fig.8).

Figure 2-8 Non-steatotic liver parenchyma treatment at 250 pulses/point, 500 pulses/point, and 1000 pulses/point demonstrating less uniform ablations. 10x magnification scale bar= 100um (A-D), 40x scale bar= 50um (E-H). Black boxes on 10x images indicate area magnified in respective 40x images. Histology images are representative of the samples treated.

Page 83: Determining the Oncological and Immunological Effects of

46

2.6 Discussion

To study the potential of histotripsy for CC tumor ablation, this work first investigated

the in vivo feasibility of histotripsy CC ablation in a PDX mouse model, with results

showing histotripsy could achieve precise and complete ablation within the targeted

region of the tumor. The treatment doses used to treat the CC tumors in this study were

higher than those reported previously for xenografted HCC tumors [28]. Given the

known correlation between tissue mechanical properties and susceptibility to histotripsy,

we hypothesize that this difference is likely due to the higher mechanical strength of CC

tumors [31-33]. Overall, the results from this in vivo feasibility study suggest that

histotripsy will be capable of treating CC tumors if the proper treatment dose is applied.

In the second part of this study, the effectiveness of histotripsy for ablating CC tumors

was compared to HCC and CLM tumors, as well as LP tissue from the surgical margin,

Figure 2-9 Steatotic liver parenchyma treatment at 250 pulses/point, 500 pulses/point, and 1000 pulses/point demonstrating less uniform ablations. 10x magnification scale bar= 100um (A-D), 40x scale bar= 50um (E-H). Black boxes on 10x images indicate area magnified in respective 40x images. Arrows indicating example cells within an ablated region that appear to be intact. Histology images are representative of the samples treated.

Page 84: Determining the Oncological and Immunological Effects of

47

in ex vivo experiments using human tumor specimens. Overall, the results of these

experiments help to demonstrate the histotripsy dosimetry metrics for treating CC in

comparison to HCC and CLM liver tumors. More specifically, the results showed that

both the CC and CLM tumors required higher treatment doses before tissue ablation

was observed in comparison to the softer HCC and LP samples that showed complete

tumor ablation at lower treatment doses. These findings match prior studies that have

shown fibrous tissues, which have been reported to have higher mechanical strength,

are more resistant to histotripsy and therefore require higher treatment doses [31-33].

Results from HCC treatments demonstrated that HCC can be completely ablated with

histotripsy with less than 500 pulses/point, agreeing with previous reports that

histotripsy has the potential for rapid, precise, and complete ablation of primary liver

tumors. Since CLM tumors are stiffer than HCC tumors [31, 32], we hypothesized that

higher treatment doses would be required for generating ablation in these samples.

Results supported this hypothesis showing histotripsy was able to achieve only small

regions of partial ablation for 500 pulses/point. At higher treatment doses of 1500 and

2000 pulses/point, nearly complete or complete ablation was observed in several CLM

samples, with the only untreated regions corresponding to highly fibrous regions within

the tumor sample.

For ex vivo experiments with CC tumors, the trends were similar to those

observed for CLM with higher doses again being needed to generate ablation in the CC

tumors. Even at the highest doses treated in this study (4000 pulses/point), there were

no cases in which we observed complete ablation of the entire treatment volume within

the CC tumors, suggesting that higher doses will be needed to achieve full ablation of

Page 85: Determining the Oncological and Immunological Effects of

48

these highly fibrous CC tumors. This finding appeared to be at least partially due to the

large variation in tissue composition between samples as well as the heterogeneity of

the tissue within a given sample (Fig.S1). Partial ablations were more pronounced in

the CC and CLM tumor samples. We hypothesize that this is due to the higher

mechanical strength and the more fibrous nature of these tumors.

Although complete ablation was not generated in the ex vivo CC tumors,

significant ablation was observed for treatments at the two highest doses tested, with

nearly 75% of the treated volume ablated after 2000 and 4000 pulses/point. Overall,

these results highlight the need for tumor-specific treatment strategies in future clinical

treatments of liver tumors with histotripsy to ensure complete ablation of CLM and CC

tumors and more efficient ablation of HCC tumors. For instance, these approaches

could include the development of guidelines for the treatment doses (histotripsy

pulses/point) needed for treating different tumor types as well as the development of

optimized pulsing parameters, although further work would be needed in order to

investigate this possibility. Additionally, improved real-time feedback methods could be

utilized to ensure complete therapeutic ablation of specific tumors such as those

previously developed using bubble-induced color doppler feedback [45, 46] or cavitation

detection [47].

In addition to the differences in ablation observed between tumor types, another

important observation from this study was the variability in ablation between samples of

a given tumor type. This result was possibly due in part to variations in the tissue

composition, mainly fibrotic deposits, between any two tumors of the same type as well

as the heterogeneity of the tissue within a single tumor. This effect would explain why

Page 86: Determining the Oncological and Immunological Effects of

49

the more uniform and softer HCC samples consistently resulted in complete ablation at

lower treatment doses compared to more variable results and less complete ablation

observed in the more heterogeneous and fibrous CLM and CC tumors. Looking at the

regions of partial ablation in these samples, results showed that the foci of ablation

interspersed within regions that weren’t ablated corresponded to the areas with less

fibrosis. This result suggests that there is a need for ensuring histotripsy treatment

doses are either optimized for each patient based on pretreatment assessments of the

tumor composition and mechanical character, such as through MRI or ultrasound

elastography. Alternatively, a sufficient treatment dose could be identified based on the

most fibrous tumors within each type based upon historically determined mechanical

properties. For example, it is possible that the different subtypes of intrahepatic CC

(mass-forming, periductal, intraductal) may respond differently to histotripsy both in the

required treatment dose as well as the efficacy of histotripsy for achieving a desired

clinical outcome. Future work should explore this possibility in order to establish the

optimal role of histotripsy for the treatment of each subtype of intrahepatic CC, as well

as extrahepatic CC, and determine if tumor type specific treatment methods are

required.

However, although we believe the variations in tissue composition are likely to

explain some of the variability in our ex vivo tissue experiments and should be

considered in future clinical histotripsy treatments, it is also important to note that these

findings could also be due to the limitations of our tissue collection methods in this

study. Unfortunately, due to the requirements of the clinical pathologists before tissue

collection, the samples collected for this study were often smaller than the desired size

Page 87: Determining the Oncological and Immunological Effects of

50

for performing optimal ex vivo histotripsy experiments. In these smaller samples,

prefocal bubble clouds were more often observed on the tissue-gel interface especially

in the more fibrous CLM and CC tumors, preventing a more uniform histotripsy

treatment from being applied to the tissue volume and likely contributing to some of the

variable ablation results observed in those tissues. These effects are not expected to

occur in vivo where much larger tumors that are surrounded by adjacent LP will be

treated with histotripsy, therefore minimizing the risk of prefocal cavitation that causes

inconsistent tumor ablation.

Another observation from the ex vivo experiments conducted in this work was the

finding that non-steatotic LP was readily ablated with histotripsy, similar to HCC

samples, whereas higher treatment doses were required to ablate steatotic LP samples.

This finding is important for future liver cancer treatments with histotripsy since it is a

standard clinical practice to ablate a margin of the surrounding liver around the tumor to

destroy all microscopic foci of tumor cells that might be within these regions. The finding

that the LP surrounding tumors in patients with hepatic steatosis, or fatty liver, required

higher treatment doses compared to those without this disease, should be noted in

future studies optimizing treatment parameters for hepatic histotripsy. It is also possible

that fatty pockets dispersed throughout the liver could contribute to more diffuse or

prefocal cavitation. This effect would make sense since previous work has shown a

significantly lower cavitation threshold for fat compared to water-based parenchymal

tissues [20, 21]. However, since no consistent differences in cavitation were observed

between the steatotic and non-steatotic LP samples treated in this study, further work

remains needed to investigate the role that fat deposits play in hepatic histotripsy and

Page 88: Determining the Oncological and Immunological Effects of

51

whether any differences in the cavitation threshold can cause variation in ablation

completeness similar to what has been observed for differences in tissue fibrosis.

2.7 Conclusion

This study tested the potential of histotripsy for the treatment of CC tumors for the

first time, with results suggesting that histotripsy has the potential to be used as a non-

invasive ablation method for the treatment of CC. Results from the in vivo experiments

demonstrated that histotripsy could generate precise and complete ablation of CC

tumors in a patient-derived xenograft CC mouse model. In the second part of this work,

which compared the histotripsy treatment dose required to ablate different types of liver

tumors, results showed that CC and CLM tumors required significantly higher doses

than HCC tumors. These findings highlight the need for tumor-specific treatment

strategies for future histotripsy liver cancer procedures to ensure more fibrous tumor

types, as well as more fibrous regions within a tumor, are completely ablated during

histotripsy. Overall, the findings of this study support further investigation of histotripsy

for the treatment of CC in addition to HCC and CLM.

2.8 Acknowledgement

This research was supported by a grant from the Focused Ultrasound

Foundation (FUSF-RAP-S-18-00011). The authors would also like to thank the ICTAS

Center for Engineered Health, the Virginia Tech Department of Biomedical Engineering

Page 89: Determining the Oncological and Immunological Effects of

52

and Mechanics, Virginia Tech Carilion School of Medicine, and the Virginia-Maryland

College of Veterinary Medicine for their support of this study. This work was conducted

in memory of Andrew J. Lockhart, who passed away from cholangiocarcinoma on

September 30, 2016. The authors encourage anyone interested to consider a donation

to the Focused Ultrasound Foundation in Andrew’s name.

Page 90: Determining the Oncological and Immunological Effects of

53

2.9 References

[1] ACS, "Cancer Facts & Figures 2020," American Cancer Society, 2020.

[2] M. C. Jansen, R. van Hillegersberg, R. A. Chamuleau, O. M. van Delden, D. J.

Gouma, and T. M. van Gulik, "Outcome of regional and local ablative therapies

for hepatocellular carcinoma: a collective review," Eur J Surg Oncol, vol. 31, no.

4, pp. 331-47, May 2005, doi: 10.1016/j.ejso.2004.10.011.

[3] L. Rossi et al., "Current approach in the treatment of hepatocellular carcinoma,"

World J Gastrointest Oncol, vol. 2, no. 9, pp. 348-59, Sep 15 2010, doi:

10.4251/wjgo.v2.i9.348.

[4] D. M. Nagorney, J. H. Donohue, M. B. Farnell, C. D. Schleck, and D. M. Ilstrup,

"Outcomes after curative resections of cholangiocarcinoma," Archives of surgery,

vol. 128, no. 8, pp. 871-878, 1993.

[5] K. Feng et al., "A randomized controlled trial of radiofrequency ablation and

surgical resection in the treatment of small hepatocellular carcinoma," Journal of

hepatology, vol. 57, no. 4, pp. 794-802, Oct 2012, doi:

10.1016/j.jhep.2012.05.007.

[6] M. S. Chen et al., "A prospective randomized trial comparing percutaneous local

ablative therapy and partial hepatectomy for small hepatocellular carcinoma," (in

eng), Ann Surg, Randomized Controlled Trial, Research Support, Non-U.S. Gov't

vol. 243, no. 3, pp. 321-8, Mar 2006, doi: 10.1097/01.sla.0000201480.65519.b8.

Page 91: Determining the Oncological and Immunological Effects of

54

[7] C. W. Hammill, K. G. Billingsley, M. A. Cassera, R. F. Wolf, M. B. Ujiki, and P. D.

Hansen, "Outcome after laparoscopic radiofrequency ablation of technically

resectable colorectal liver metastases," (in eng), Annals of surgical oncology, vol.

18, no. 7, pp. 1947-54, Jul 2011, doi: 10.1245/s10434-010-1535-9.

[8] G. Otto, C. Duber, M. Hoppe-Lotichius, J. Konig, M. Heise, and M. B. Pitton,

"Radiofrequency ablation as first-line treatment in patients with early colorectal

liver metastases amenable to surgery," Ann Surg, vol. 251, no. 5, pp. 796-803,

May 2010, doi: 10.1097/SLA.0b013e3181bc9fae.

[9] R. Salem et al., "Institutional Decision to Adopt Y90 as Primary Treatment for

HCC Informed by a 1,000-patient 15-year Experience," Hepatology, Dec 1 2017,

doi: 10.1002/hep.29691.

[10] A. Forner, J. M. Llovet, and J. Bruix, "Hepatocellular carcinoma," Lancet, vol.

379, no. 9822, pp. 1245-55, Mar 31 2012, doi: 10.1016/S0140-6736(11)61347-0.

[11] L. European Association for the Study of the, R. European Organisation for, and

C. Treatment of, "EASL–EORTC Clinical Practice Guidelines: Management of

hepatocellular carcinoma," Journal of hepatology, vol. 56, no. 4, pp. 908-943,

2012. [Online]. Available:

http://linkinghub.elsevier.com/retrieve/pii/S0168827811008737?showall=true.

[12] A. Gillams et al., "Thermal ablation of colorectal liver metastases: a position

paper by an international panel of ablation experts, The Interventional Oncology

Sans Frontieres meeting 2013," Eur Radiol, vol. 25, no. 12, pp. 3438-54, Dec

2015, doi: 10.1007/s00330-015-3779-z.

Page 92: Determining the Oncological and Immunological Effects of

55

[13] N. Rozenblum et al., "Oncogenesis: An "Off-Target" Effect of Radiofrequency

Ablation," Radiology, vol. 276, no. 2, pp. 426-32, Aug 2015, doi:

10.1148/radiol.2015141695.

[14] B. Sangro et al., "Prevention and treatment of complications of selective internal

radiation therapy: Expert guidance and systematic review," Hepatology, vol. 66,

no. 3, pp. 969-982, Sep 2017, doi: 10.1002/hep.29207.

[15] C. M. Smolock AR, Vlaisavljevich E, Gendron-Fitzpatrick A, Green C, Ziemlewicz

TJ and Lee FT, "Robotically Assisted Sonic Therapy as a Non-Invasive Non-

Thermal Ablation Modality: Proof of Concept in a Porcine Liver Model,"

Radiology, vol. 287, no. 2, pp. 485-93, 2018.

[16] S. X. Vidal Jove J, Vlaisavljevich E, Cannata J, Duryea A, Miller R, Lee F and

Ziemlewicz T, "Phase I Study of Safety and Efficacy of Hepatic Histotripsy:

Preliminary Results of First in Man Experience with Robotically-Assisted Sonic

Therapy," in International Society of Therapeutic Ultrasound, Barcelona, Spain,

2019.

[17] E. Vlaisavljevich et al., "Non-Invasive Ultrasound Liver Ablation Using Histotripsy:

Chronic Study in an In Vivo Rodent Model," Ultrasound Med Biol, vol. 42, no. 8,

pp. 1890-902, Aug 2016, doi: 10.1016/j.ultrasmedbio.2016.03.018.

[18] E. Vlaisavljevich et al., "Image-guided non-invasive ultrasound liver ablation

using histotripsy: feasibility study in an in vivo porcine model," Ultrasound Med

Biol, vol. 39, no. 8, pp. 1398-409, Aug 2013, doi:

10.1016/j.ultrasmedbio.2013.02.005.

Page 93: Determining the Oncological and Immunological Effects of

56

[19] T. Worlikar et al., "Histotripsy for Non-Invasive Ablation of Hepatocellular

Carcinoma (HCC) Tumor in a Subcutaneous Xenograft Murine Model," Conf Proc

IEEE Eng Med Biol Soc, vol. 2018, pp. 6064-6067, Jul 2018, doi:

10.1109/EMBC.2018.8513650.

[20] A. D. Maxwell, C. A. Cain, T. L. Hall, J. B. Fowlkes, and Z. Xu, "Probability of

cavitation for single ultrasound pulses applied to tissues and tissue-mimicking

materials," Ultrasound Med Biol, vol. 39, no. 3, pp. 449-65, Mar 2013, doi:

10.1016/j.ultrasmedbio.2012.09.004.

[21] E. Vlaisavljevich et al., "Effects of ultrasound frequency and tissue stiffness on

the histotripsy intrinsic threshold for cavitation," Ultrasound Med Biol, vol. 41, no.

6, pp. 1651-67, Jun 2015, doi: 10.1016/j.ultrasmedbio.2015.01.028.

[22] Z. Xu et al., "Controlled ultrasound tissue erosion," IEEE Trans Ultrason

Ferroelectr Freq Control, vol. 51, no. 6, pp. 726-36, Jun 2004, doi:

10.1109/tuffc.2004.1308731.

[23] R. M. Xu Z, Hall TL, Mycek M-A, Fowlkes JB and Cain CA, "Evolution of bubble

clouds produced in pulsed cavitational ultrasound therapy - histotripsy," IEEE

Trans Ultrason Ferroelectr Freq Control, vol. 55, pp. 1122-32, 2008.

[24] E. Vlaisavljevich, A. Maxwell, M. Warnez, E. Johnsen, C. A. Cain, and Z. Xu,

"Histotripsy-induced cavitation cloud initiation thresholds in tissues of different

mechanical properties," IEEE Trans Ultrason Ferroelectr Freq Control, vol. 61,

no. 2, pp. 341-52, Feb 2014, doi: 10.1109/TUFFC.2014.6722618.

[25] E. Vlaisavljevich, A. Maxwell, L. Mancia, E. Johnsen, C. Cain, and Z. Xu,

"Visualizing the Histotripsy Process: Bubble Cloud-Cancer Cell Interactions in a

Page 94: Determining the Oncological and Immunological Effects of

57

Tissue-Mimicking Environment," Ultrasound Med Biol, vol. 42, no. 10, pp. 2466-

77, Oct 2016, doi: 10.1016/j.ultrasmedbio.2016.05.018.

[26] V. E. Kim Y, Owens GE, Allen SP, Cain CA and Xu Z, "In vivo transcostal

histotripsy therapy without aberration correction," Phys Med Biol, vol. 59, pp.

2553-68, 2014.

[27] E. Vlaisavljevich et al., "Non-Invasive Liver Ablation Using Histotripsy: Preclinical

Safety Study in an In Vivo Porcine Model," Ultrasound Med Biol, vol. 43, no. 6,

pp. 1237-1251, Jun 2017, doi: 10.1016/j.ultrasmedbio.2017.01.016.

[28] T. Worlikar et al., "Histotripsy for Non-Invasive Ablation of Hepatocellular

Carcinoma (HCC) Tumor in a Subcutaneous Xenograft Murine Model," in 2018

40th Annual International Conference of the IEEE Engineering in Medicine and

Biology Society (EMBC), 2018: IEEE, pp. 6064-6067.

[29] S. Qu et al., "Non-thermal histotripsy tumor ablation promotes abscopal immune

responses that enhance cancer immunotherapy," Journal for ImmunoTherapy of

Cancer, vol. 8, no. 1, 2020.

[30] S. K. Venkatesh et al., "MR elastography of liver tumors: preliminary results,"

American Journal of Roentgenology, vol. 190, no. 6, pp. 1534-1540, 2008.

[31] E. Vlaisavljevich, Y. Kim, G. Owens, W. Roberts, C. Cain, and Z. Xu, "Effects of

tissue mechanical properties on susceptibility to histotripsy-induced tissue

damage," (in eng), Phys Med Biol, vol. 59, no. 2, pp. 253-70, Jan 20 2014, doi:

10.1088/0031-9155/59/2/253.

[32] E. Vlaisavljevich, A. Maxwell, M. Warnez, E. Johnsen, C. Cain, and Z. Xu,

"Histotripsy-induced cavitation cloud initiation thresholds in tissues of different

Page 95: Determining the Oncological and Immunological Effects of

58

mechanical properties," (in eng), IEEE Transactions on Ultrasonics,

Ferroelectrics, and Frequency Control, vol. 61, no. 2, pp. 341-52, Feb 2014, doi:

10.1109/TUFFC.2014.6722618.

[33] E. Vlaisavljevich, Z. Xu, A. Arvidson, L. Jin, W. Roberts, and C. Cain, "Effects of

thermal preconditioning on tissue susceptibility to histotripsy," Ultrasound in

medicine & biology, vol. 41, no. 11, pp. 2938-2954, 2015.

[34] A. Guibal et al., "Evaluation of shearwave elastography for the characterisation of

focal liver lesions on ultrasound," European radiology, vol. 23, no. 4, pp. 1138-

1149, 2013.

[35] T. G. Schuster, J. T. Wei, K. Hendlin, R. Jahnke, and W. W. Roberts, "Histotripsy

Treatment of Benign Prostatic Enlargement Using the Vortx Rx System: Initial

Human Safety and Efficacy Outcomes," Urology, vol. 114, pp. 184-187, Apr

2018, doi: 10.1016/j.urology.2017.12.033.

[36] E. Vlaisavljevich et al., "Non-invasive liver ablation using histotripsy: preclinical

safety study in an in vivo porcine model," Ultrasound in medicine & biology, vol.

43, no. 6, pp. 1237-1251, 2017.

[37] W. W. Roberts, D. Teofilovic, R. C. Jahnke, J. Patri, J. M. Risdahl, and J. A.

Bertolina, "Histotripsy of the prostate using a commercial system in a canine

model," The Journal of urology, vol. 191, no. 3, pp. 860-865, 2014.

[38] A. D. Maxwell et al., "Cavitation clouds created by shock scattering from bubbles

during histotripsy," J Acoust Soc Am, vol. 130, no. 4, pp. 1888-98, Oct 2011, doi:

10.1121/1.3625239.

Page 96: Determining the Oncological and Immunological Effects of

59

[39] J. E. Parsons, C. A. Cain, G. D. Abrams, and J. B. Fowlkes, "Pulsed cavitational

ultrasound therapy for controlled tissue homogenization," (in eng), Ultrasound in

Medicine and Biology, vol. 32, no. 1, pp. 115-29, Jan 2006, doi: S0301-

5629(05)00374-1 [pii]

10.1016/j.ultrasmedbio.2005.09.005.

[40] J. E. Parsons, C. A. Cain, and J. B. Fowlkes, "Cost-effective assembly of a basic

fiber-optic hydrophone for measurement of high-amplitude therapeutic ultrasound

fields," J Acoust Soc Am, vol. 119, no. 3, pp. 1432-40, Mar 2006, doi:

10.1121/1.2166708.

[41] E. Vlaisavljevich, T. Gerhardson, T. Hall, and Z. Xu, "Effects of f-number on the

histotripsy intrinsic threshold and cavitation bubble cloud behavior," Phys Med

Biol, vol. 62, no. 4, pp. 1269-1290, Feb 21 2017, doi: 10.1088/1361-

6560/aa54c7.

[42] R. M. Brock et al., "Patient derived xenografts expand human primary pancreatic

tumor tissue availability for ex vivo irreversible electroporation testing," Frontiers

in Oncology, vol. 10, 2020.

[43] K. C. Longo et al., "Histotripsy Ablations in a Porcine Liver Model: Feasibility of

Respiratory Motion Compensation by Alteration of the Ablation Zone Prescription

Shape," CardioVascular and Interventional Radiology, pp. 1-7, 2020.

[44] E. Vlaisavljevich, Y. Kim, G. Owens, W. Roberts, C. Cain, and Z. Xu, "Effects of

tissue mechanical properties on susceptibility to histotripsy-induced tissue

damage," Physics in Medicine & Biology, vol. 59, no. 2, p. 253, 2013.

Page 97: Determining the Oncological and Immunological Effects of

60

[45] J. J. Macoskey et al., "Bubble-induced color Doppler feedback correlates with

histotripsy-induced destruction of structural components in liver tissue,"

Ultrasound in medicine & biology, vol. 44, no. 3, pp. 602-612, 2018.

[46] T. Y. Wang, T. L. Hall, Z. Xu, J. B. Fowlkes, and C. A. Cain, "Imaging feedback of

histotripsy treatments using ultrasound shear wave elastography," (in eng), IEEE

Transactions on Ultrasonics, Ferroelectrics, and Frequency Control, Research

Support, N.I.H., Extramural vol. 59, no. 6, pp. 1167-81, Jun 2012, doi:

http://dx.doi.org/10.1109/TUFFC.2012.2307.

[47] J. J. Macoskey et al., "Using the cavitation collapse time to indicate the extent of

histotripsy-induced tissue fractionation," (in eng), Phys Med Biol, vol. 63, no. 5, p.

055013, Mar 8 2018, doi: 10.1088/1361-6560/aaae3b.

Page 98: Determining the Oncological and Immunological Effects of

61

Chapter 3. Histotripsy for Non-Invasive Ablation of Cholangiocarcinoma Carcinoma in a Patient-Derived Xenograft Model

Alissa Hendricks-Wenger, Sofie Saunier, Alexander Simon, Sheryl Coutermarsh-Ott1, Irving C. Allen, Eli Vlaisavljevich

This chapter is excerpted from a manuscript in preparation for submission to Ultrasound

in Medicine and Biology.

Page 99: Determining the Oncological and Immunological Effects of

62

3.1 Abstract

Histotripsy is a non-invasive focused ultrasound ablation method that is currently

being developed for the treatment of primary and metastatic liver tumors. In a recent

study, we investigated the feasibility of using histotripsy for the ablation of

cholangiocarcinoma (CC), a rare form of liver cancer of the bile duct that is difficult to

treat with current therapies due to its common location near critical structures, late

diagnosis, and fibrous tissue microenvironment. As part of this prior study, the in vivo

feasibility of treating CC tumors was show in an acute patient-derived xenografted

(PDX) CC mouse model. Here, we investigate histotripsy for the ablation of CC tumors

in vivo in a chronic PDX mouse model of CC. Patient-derived xenografted CC mice

were treated with histotripsy using a 1MHz transducer at a PRF of 500 Hz and an

applied dose of 500 pulses/point. First, a pilot study was conducted to determine if

histotripsy could safely treat the entire CC tumors plus a ~2mm surgical margin in the

PDX model without inducing significant adverse events. Results showed histotripsy

generated significant injuries to intestinal tissues located within the treatment margin,

precluding the use of this positive-margin treatment strategy in subsequent

experiments. For the next set of experiments, histotripsy was applied to CC tumors

(n=6) with the bubble cloud contained only to the targeted tumor (zero-margin), resulting

in ~80%-90% of the tumor treated and no cavitation events outside of the tumor. After

treatment, ablation was visualized using ultrasound imaging immediately post-treatment

and subjects were then monitored over time to determine changes in tumor size, long-

Page 100: Determining the Oncological and Immunological Effects of

63

term survival, and safety outcomes assessed by adverse events. Results were

compared to control mice that did not receive histotripsy (n=6) and showed histotripsy

achieved an average of 73% reduction of tumor diameter 26 days after treatment, with

no significant adverse events or damage to surrounding tissues. Notably, 3 of the 6

treated tumors were reduced in diameter to an undetectable size after ~2.5 weeks as

the ablated homogenate was reabsorbed, before tumor growth was again observed.

The treated animals were found to have significantly increased progression-free and

overall survival, and no metastases were observed in any animals. Overall, results

support the hypothesis that histotripsy can be used as a safe and effect method for

treating CC tumors while also highlighting the limitations of the subcutaneous murine

model.

3.2 Keywords

Focused Ultrasound, Patient Derived Xenograft, Cholangiocarcinoma, Tumor Ablation,

Histotripsy

Page 101: Determining the Oncological and Immunological Effects of

64

3.3 Introduction

Cholangiocarcinoma (CC) is a rare form of liver cancer that affects ~8,000 people in

the United States each year and has a 5-year relative survival rate of only 8% [1, 2].

Current treatment options include surgery, chemoembolization, and radiotherapy;

however, due to their location near vital structures, CC tumors pose a unique challenge

for these modalities [3-5]. For example, although curative resection offers the best

chance for long-term survival, only a fraction of CC tumors are considered to be

resectable, including 15% of intrahepatic CC tumors and 56% of extrahepatic CC

tumors [6]. There has been some success in using thermal ablation methods for the

treatment of the primary liver tumors such as hepatocellular cholangiocarcinoma as well

as metastatic liver tumors. However, the lack of precision and high complication rates

when in proximity to critical structures present significant limitations for thermal ablation

[7-14]. Due to this limitation, thermal ablation is often ruled out as a treatment option for

CC due to the high risk of complications for treating CC tumors which are most often

located in close proximity to the bile ducts [14].

Histotripsy is an image-guided, non-invasive, non-thermal, and non-ionizing tumor

ablation method that uses focused ultrasound to ablate targeted tissue through

controlled generation of acoustic cavitation [15-17]. Histotripsy delivers focused,

microsecond long, high-pressure ultrasound pulses that generate a cavitation “bubble

cloud” inside of the target tissue at the focus of the transducer [18-22]. The generation

of these cavitation bubbles, which rapidly expand and collapse, induces high strain to

Page 102: Determining the Oncological and Immunological Effects of

65

cells within the tumor and results in precise, non-thermal tissue ablation [23]. Histotripsy

has shown great promise for the treatment of liver cancer in both preclinical studies [15,

16, 24-28] and early clinical trials [29]. These studies have shown that histotripsy can

safely and non-invasively ablate tumors in the liver with high precision, including regions

located near critical structures such as bile ducts, nerves, and blood vessels [22, 30-36].

More specifically, due to its non-thermal nature, histotripsy has shown the ability to

effectively ablate tissue surrounding these critical structures without causing damage to

their anatomy, likely due to the higher mechanical strength and fibrotic composition of

these structures [16, 22, 27, 31, 35].

Due to these previous studies showing the potential of hepatic histotripsy, our team

recently completed a feasibility study to investigate whether histotripsy could be used

for the ablation of CC tumors [37]. The ex vivo portion of this study demonstrated that

CC tumors require a higher dose (i.e. more histotripsy pulses per treatment point) to

reach comparable ablation qualities to other liver malignancies, specifically

hepatocellular carcinoma and colorectal carcinoma tumors that metastasized to the

liver. The higher treatment dose required for CC tumors was hypothesized to be due to

the higher mechanical strength and more fibrotic composition of the CC tumors in

comparison to hepatocellular carcinoma and metastatic liver tumors [37, 38]. This

previous study also investigated the in vivo feasibility of histotripsy for treating CC

tumors in a patient-derived xenograft (PDX) CC tumor model, with results showing

histotripsy could achieve ablation of CC tumors using a 1MHz small animal histotripsy

transducer [17, 37]. Here, we build on this prior work in order to investigate the safety

and efficacy of histotripsy for treating CC tumors in the subcutaneous PDX mouse

Page 103: Determining the Oncological and Immunological Effects of

66

model of CC. This model was chosen due the tumor-specific tumor microenvironment

that will allow for proper assessment of tumor ablation success [39] as well as the lack

of other well-established orthotopic murine or large animal CC tumor models. In the first

part of this study, we investigated whether histotripsy could be used to safely generate

complete tumor ablation plus a positive surgical margin in the PDX tumor model. Based

on the results from these pilot studies, a second set of experiments was conducted in

order to investigate the long-term response of CC tumors treated with histotripsy using

more conservative treatments in which the histotripsy bubble cloud was contained

inside the tumor, with these zero-margin treatments resulting in ~80-90% of the tumor

volume targeted with histotripsy in order to avoid off target damage to surrounding

tissues. The acute and chronic response of the murine PDX tumors were evaluated to

determine the safety and feasibility of treating CC tumors with histotripsy through

imaging, heath monitoring, and histological analysis.

3.4 Materials and Methods

3.4.1 Tumor Injections and Health Monitoring

Seventeen female NSG mice with subcutaneous flank patient derived

xenografted (PDX) CC tumors were ordered from and prepared by Jackson Laboratory

(Bar Harbor, Maine, USA). Through a process previously described for pancreatic PDX

tumors [39], a human CC tumor was collected from a patient and propagated as a

heterogeneous tumor in immunocompromised NSG mice. The PDX tumor (TM01225)

Page 104: Determining the Oncological and Immunological Effects of

67

was a diagnosed adenosquamous carcinoma, a subtype of CC, with an unspecified

grade/stage that was surgically resected from the bile duct, the primary site, of a 79-

year-old white/Hispanic male. Mice were delivered to our facility after confirmed tumor

engraftment. After arrival, animals were monitored for general health, weight, and tumor

size at least 3 times per week until tumors reached the desired treatment size of ~0.4cm

in diameter. Tumor diameters were measured with calipers and calculated as the

square root of two perpendicular measurements, as previously described [39]. All

animal experiments were approved and carried out in accordance with the Virginia Tech

Institutional Animal Care and Use Committee under IACUC protocol 18-031-CVM.

3.4.2 Histotripsy Setup

The in vivo histotripsy treatment was applied using a custom 8-element, 1 MHz

small animal histotripsy transducer with a geometric focus of 36 mm, an aperture size of

52.7 mm, and a f-number of 0.68. The full-width half-maximum dimensions at the

geometric focus of this transducer were 0.98 mm, 0.93 mm, and 3.9 mm in the

transverse, elevational, and axial directions, respectively. A custom high-voltage pulser

that generates short therapy pulses of <2 cycles was used to drive the transducer, while

being controlled by a field-programmable gate array (FPGA) board (Altera DE0-Nano

Terasic Technology, Dover, DE, USA) programmed for histotripsy therapy pulsing and

powered by a high voltage DC power supply (GENH750W, TDK-Lambda). During

treatment, the transducer was positioned underneath a custom-designed surgical

mouse stage (Fig.1A) ensuring its submersion in the tank of degassed water. A

Page 105: Determining the Oncological and Immunological Effects of

68

computer-guided positioning system on a 3-D axis with a 0.05 mm motor resolution was

attached to the transducer in order to run the automated volumetric treatments using a

custom user-interface operated through MATLAB. A linear ultrasound imaging probe

with a frequency range from 10-18 MHz (L18-10L30H-4, Telemed, Lithuania, EU) was

coaxially aligned within the transducer for real-time treatment guidance and monitoring.

This treatment set-up has previously been used for treating subcutaneous and

orthotopic tumors with histotripsy in small animal models [15, 17, 40-43], including our

recent study testing the acute feasibility of ablating CC tumors in the PDX CC tumor

model [37].

3.4.3 Histotripsy Treatment Procedures

Mice were treated when the average cohort tumor diameter was approximately

0.4cm in diameter, to ensure that the smallest tumors were large enough for targeting,

at least 0.3cm in the shortest diameter. Prior to treatment, the water tanks were filled

and degassed, and a water heater was used to maintain the water temperature at

37+4oC. The area of the mouse flank that contained the tumors had fur removed by

applying Nair (Naircare, Ewing, NJ) to the area for 2-4 minutes then washing the area

clean with water and paper towels. Mice were anesthetized with vaporized isoflurane (1-

3% with 1.5 L/min O2 flow rate) and the tumor was submerged in the water through an

opening in the surgical stage. Ultrasound guidance was used to locate the tumor and

confirm the location of the bubble cloud within the tumor throughout the duration of the

treatments (Fig.1C). Using the automated volumetric code to control treatments, an

Page 106: Determining the Oncological and Immunological Effects of

69

ellipsoid was overlaid onto the tumor, and divided into disks set 0.25mm apart. Points

within each of these disks were set 0.25 mm apart across the entire tumor and 0.5 mm

apart through the depth in the axial direction (Fig.1B). Each point in the disks was

treated with a dose of 500 histotripsy pulses using a pulse repetition frequency (PRF) of

500 Hz and a dwell time of 1 second. The treatment was programmed to move from the

center disk to the external disk, and the same process was then repeated for the

second half of the tumor. For each disk, the treatment started at the central point and

then raster scanned one half and then returned to the center for the second half, to

ensure maximum ablative efficacy.

3.4.4 Pilot Studies to Confirm Ablation Dose and Treatment Margins

Two pilot studies were conducted in order to establish the treatment dose and

margins to be used for chronic experiments. First, negative margin treatments (Fig.2)

were used in an acute pilot study to reconfirm that the dose determined in our prior CC

PDX tumor study was appropriate for this cohort of PDX tumors (n=3) [37]. For these

animals, euthanasia was preformed within 30 minutes after treatment. To establish the

potential of a complete tumor ablation, a second pilot study was conducted testing

positive-margin treatments (Fig.2) which included a ~1-2mm positive margin around the

tumor (n=2) regardless of inclusion of off-target structures. To minimize damage to off

target-tissues (i.e. muscles, skin, and intestines), the therapy transducer was manually

turned off for any bubble cloud that was visually found on ultrasound imaging to be

100% outside of the tumor margins during treatment. Animals in this group were

Page 107: Determining the Oncological and Immunological Effects of

70

allowed to recover from treatment and were euthanized when it was determined that

pain thresholds were reached with no signs of improvement after subsequent 4-hour

checks, as guided by our IACUC protocol. The symptoms evaluated for these

assessments included lethargy, decreased socialization, difficulty rearing, decreased

extremity strength, squinting, ruffling, as well as irregular respiratory rate and rhythm.

Figure 3-1 Rodent system treatment set up (A). Ultrasound imaging confirmed presence of bubble cloud (indicated by the orange arrow) within CC tumors during treatment (B). For treatment automation, each tumor was defined as a spheroid divided into disks (C). Each treatment disk consisted of a series of points that were used for raster scanning (D). White arrow in (D) represents the direction of therapeutic ultrasound wave propagation.

Page 108: Determining the Oncological and Immunological Effects of

71

3.4.5 Survival Study

A survival study was conducted to compare histotripsy ablation of CC tumors

between treated and untreated animals (n=6 each). For the survival study, treatment

was applied using a zero-margin method to achieve as close to 100% of the tumor

being targeted with histotripsy as possible while avoiding internal structures such as

intestines being treated around the tumor as was observed in the positive-margin pilot

studies. This zero-margin treatment (Fig.2) led to approximately ~80-90% of the tumor

being targeted. Due tumor shape irregularities, swelling during treatment, and minor

animal movements, some variation was observed in the extent of tumor ablation across

the 6 treated animals. To minimize off target damage, the therapy was manually turned

off for points at which the bubble cloud was visualized to form completely outside of the

tumor. Given the margins used in this cohort, this did not occur often. After treatment,

the animals were recovered and monitored for health responses over the following

weeks. Animals were euthanized when they reached survival endpoints which consisted

of tumors exceeding 1.4 cm in calculated diameter; debilitating health noted including

irregular respiratory rate and rhythm, decreased activity, decreased socialization, or

difficulty moving extremities; or when the IACUC approved end of study time point of 60

days was reached. Progression free survival was defined as growing beyond 20% of the

average tumor diameter that was measured on the day of treatment.

Page 109: Determining the Oncological and Immunological Effects of

72

Figure 3-2 Schematic showing the planned ablation volumes treatments utilized for this study. Negative-margin treatments were set to have the entire treatment contained within the tumor; zero-margin treatments were set to treat as close to 100% of the tumor as possible without including a significant amount of non-tumor tissue; and positive-margin treatments were set to treat the entire tumor along with sufficient margin that encompassed enough non-tumor tissue to ensure complete ablation of the entire tumor. Arrows indicate tumor tissue not targeted by each planned ablation zone while * indicate intestinal tissue within the treatment margin. Figure created using Biorender.com.

3.4.6 Necropsy and Histopathology

All animals in the positive-margin pilot study and the survival study were

euthanized and necropsied as the animals individually reached the survival end points

described above. During necropsy, all tissues within abdominal cavity as well as nearby

skin and muscles were examined for abnormalities that could have been caused by

treatment, including but not limited to bruising, lacerations, swelling, and deformities.

The tumor, lungs, and liver were collected from all animals and fixed in 10% formalin.

These tissues were then paraffin-embedded and stained with hematoxylin and eosin

(H&E) following standard protocols. Evaluations of tumor tissue to evaluate treatment as

well as the lungs and liver for metastasis were performed by trained individuals and

independently verified by a blinded, board-certified pathologist (D.G.).

Page 110: Determining the Oncological and Immunological Effects of

73

3.4.7 Statistics

Tumor growth and survival data were analyzed using GraphPad Prism, version 8.

A Student’s two-tailed t-test was used when comparing the survival study experimental

groups tumor growth. Standard Kaplan-Meyer analysis was conducted on progression

free and overall survival. Progression free survival was defined as the tumor exceeding

120% the average diameter on treatment day and overall survival was determined

based on morbidity, as described above. Statistical significance was defined as p ≤ 0.05.

Data are represented as the mean +/- SEM.

Figure 3-3 Histological examination revealed undisturbed tumor stroma in acute untreated mice (A) and completely acellular targeted regions in treated tumors (B). Comparing to before treatment (C), the hypoechoic regions were identifiable in the ultrasound imaging after treatment (D). The treatment area, represented by the dotted yellow circle, closely matched the negative-margin planned ablation volume on ultrasound imaging.

Page 111: Determining the Oncological and Immunological Effects of

74

3.5 Results

3.5.1 Pilot Studies of Ablation Dose and Margins

Figure 3-4 Using positive, more aggressive margins (n=2) there was more significant skin lesions and subcutaneous bruising that extended well beyond the treatment area across the abdomen (A). Darkened, parrelel lesions that correspond to treatment disks are emphasized with purple arrows. At necropsy, these more aggressive treatments revealed free fluid and intestinal contents within the abdomen and a clear intestinal perforation, indicated by the green arrow (B).

Histotripsy generated a well-confined bubble cloud that destroyed the targeted

CC tumor. Histology results revealed an undisturbed tumor stroma in the untreated mice

(Fig.3A) and a completely acellular region in the treated tumor (Fig.3B). Corresponding

ultrasound guidance for pre-treatment imaging showed the tumor as a hyperechoic

region (Fig.3C), and post-treatment the target region became hypoechoic visualized by

the darker region within the tumor margins (Fig.3D).

When using the positive-margin treatments (n=2), there was no palpable tumor

immediately after treatment, suggesting complete ablation of the entire tumor was

achieved. For both animals treated with the positive-margin ablation volume, significant

side effects were observed including slight bleeding during treatment and post-

treatment skin abrasions that appeared to align with the treatment disks (Fig.4A). In

addition to the skin lesions, there was notable bruising and swelling behind the

Page 112: Determining the Oncological and Immunological Effects of

75

treatment region, with bruising extending down the flank and across the abdomen.

Immediately after treatment, both animals were found to be bright, alert, and oriented.

At the 12- or 36-hour post-treatment health checks, the mice were able to react to

stimuli normally and had normal range of movement. However, at these checks due to

the increased signs of an adverse event, including lethargic behavior and showed signs

of discomfort and pain, one of the mice were euthanized at each check. Upon necropsy,

it was found that there was free blood and intestinal contents within the abdominal

cavity (Fig.4B). Closer examination found that a loop of small bowel that presided

directly behind the treatment zone within the positive treatment margin had been

perforated in one of the two mice. No other injuries were found. The tumor tissue could

not be harvested for histology from these two animals, given that it appeared to be

completely liquefied and gone at these time points.

3.5.2 Survival Study

Animals that were treated with the zero-margin histotripsy treatments (n=6)

showed no significant skin abrasions during treatment (Fig.5A). During two treatments

small lesions with slight bleeding were noted and completely resolved prior to the

completion of the treatment. Immediately after treatment, there was slight bruising in the

treated areas (Fig.5B). Three days after treatment there was no skin damage in the

untreated mice (Fig.5C) and small, pitted scabs over the treated areas in the histotripsy

treated animals (Fig.5D). Within one week of treatment, these scabs has resolved and

there were no visual differences between the treated and untreated animals. In addition,

Page 113: Determining the Oncological and Immunological Effects of

76

no signs of pain, distress, nor discomfort were observed after treatment in any of these

animals treated with the zero-margin treatments.

Figure 3-5 No significant skin breaks were observed during treatment (A). Immediately after treatment, there was slight bruising surrounding the tumors, indicated by the blue arrow (B). Three days after treatment there was no skin damage in the untreated mice (C) and small, pitted scabs over the treated areas, noted by the red arrow (D).

Tumor measurements post-treatment were taken with calipers weekly in order to

calculate the average tumor diameter reduction after treatment. Compared to the tumor

diameters in the untreated mice, the diameters of the treated tumors were significantly

reduced and growth retarded for multiple weeks post-treatment (Fig.6A), with a

maximum reduction in tumor size of 73% (p<0.05). Looking at individual mice, it can be

seen that 3 of the 6 of the treated tumors reached a point where the tumor was no

B A

D C

Page 114: Determining the Oncological and Immunological Effects of

77

longer identifiable approximately 2.5 weeks after treatment (Fig.6B). However, by the

end of the 60-day study, all mice in the treatment group had regrowth of their tumors.

Quantifying the effects of the tumor growth retardation showed that the progression free

and overall survival was significantly higher for the treated mice versus the untreated

(Fig.7). The untreated mice saw tumor growth progression on average by 21 days post

treatment and reached survival end points on average 40 days after treatment. For the

treated mice, progression of tumor growth was prevented until 40 days and survival at

day 60. Of the mice that were treated, 3 of the 6 mice did not reach survival endpoints

by the end of the study.

Figure 3-6 Average tumor diameter (mean + SEM) reduction after treatment, after treatment the difference between groups was significant at every time point (* p<0.05) (A). Individual treated and untreated mouse tumor measurements, emphasizing that multiple treated animals reached a tumor diameter of zero (B). Vertical dotted line emphasizes treatment on day 8. Horizontal dotted line at 14mm emphasizes maximum tumor diameter that any individual animal was allowed to reach in this study based on our IACUC protocols.

At survival time points, the untreated (Fig.8A) and treated (Fig.8B) tumors were

comparable in histologic state. In general, the tumors from all the mice maintained the

features that were consistent with the diagnosed adenosquamous carcinoma from the

Page 115: Determining the Oncological and Immunological Effects of

78

human donor. At the end of the study, the treated tumors had regrown, and displayed a

comparable level of fibrosis, keratin deposits, and mucin producing areas to the

untreated tumors. One notable difference between the two groups was that the

untreated tumors had larger calcium deposits within the tumors. There was no sign of

metastatic disease in the abdominal and thoracic cavities of any animal, nor did any

animal have signs of splenomegaly. For both the chronic treated and untreated mice the

liver (Fig.8C-D) and lung (Fig.8E-F) parenchyma were benign, with no gross or

histological signs of metastatic disease.

Figure 3-7 Progression free and overall survival increased (p=0.09 and 0.03 respectively) in treatment group compared to untreated. Progression free survival was defined as surpassing 120% of the average tumor diameter the day of treatment (5.29 mm). Vertical dotted line emphasizes treatment on day 8.

Page 116: Determining the Oncological and Immunological Effects of

79

Figure 3-8 Histological examination revealed undisturbed tumor stroma in chronic (A) untreated mice and tumor regrowth in chronic treated tumors (B). The liver (C-D) and lung (E-F) parenchyma in both untreated (E,C) and treated (D,F) chronic mice were found to be benign. Scale bar = 500um.

3.6 Discussion

In this study, the effects of histotripsy on the treatment of CC in an acute and

survival PDX model was investigated. Results from acute experiments showed that

Page 117: Determining the Oncological and Immunological Effects of

80

histotripsy generated defined bubble clouds that resulted in a large portion of the CC

tumors being destroyed using a treatment dose of 500 pulses/point (Fig.2), matching

our previous work establishing the feasibility of treating CC tumors with histotripsy [37].

This study was conducted in addition to our previous study that investigated the dose

needed to treat CC PDX tumors in mice with histotripsy in order to confirm this ablation

dose was effective in the new cohort of PDX mice since prior studies of PDX models

have shown significant differences in the tumor microenvironment between cohorts of

PDX mice derived from different patients [39, 44]. It is worth noting that the 500

pulses/point treatment dose used in this study was significantly higher than the 50

pulses/point dose used to achieve a similar level of tumor ablation and growth

retardation in a prior study of hepatocellular carcinoma tumors [17]. As discussed in our

previous study [37], this higher treatment dose for the CC tumors is likely due to the

higher Youngs modulus (56.9±25.6 kPa) [38] for CC tumors compared to hepatocellular

carcinoma tumors (14.86±10.0 kPa) In the prior study of histotripsy for treating

subcutaneous hepatocellular carcinoma tumors, the use of positive margins led to a

report of damage to muscles that were near the tumor [17]. Similarly, the results from

our study demonstrated that histotripsy could not be applied with positive margins

around the tumor without debilitating side effects, resulting in the need for more

conservative margins being used for the survival experiments in this study. More

specifically, results from the positive-margin treatments showed significant damage to

the intestines (Fig.3B) as well as signs of significant pain and distress after treatment. It

is likely that the higher treatment dose of 500 pulses/point needed to ablate CC tumors

increase the extent of damage to the intestines, suggesting that the tissue selective

Page 118: Determining the Oncological and Immunological Effects of

81

features of histotripsy may be less significant when treating fibrous tumors such as CC,

although further work will be needed to fully investigate this possibility. Overall, the

findings from the initial pilot studies demonstrated the need for more conservative zero-

margin treatments to be used to study the long-term effects of treating CC tumors with

histotripsy. In the animals that were treated with zero-margins, there were no negative

side effects noted beyond mild bruising and scabbing in the days immediately following

treatment (Fig.4).

Results from the survival study showed histotripsy significantly reduced tumor

burden and expanded survival measures compared to control mice. Post-treatment

measurements showed a significant and steady decrease in tumor volume compared to

the untreated groups (Fig.5) for a period of ~3.5 weeks after treatment. The treated

mice survived the histotripsy treatments when utilizing a zero-margin protocol so that

the vast majority of the tumor was treated while avoiding off-target damage, such as

bowel or intestine perforations. Given that these mice were immunocompromised and

therefore lack the immune response necessary to clear any remaining viable cells, it is

not surprising that all treated mice tumor showed tumor regrowth occurred from

surviving cancer cells in the same location as the primary tumor, as expected based on

the zero-margin treatments which did not cover 100% of the tumor volume. This

regrowth also occurred within the 3 mice that reached a point were no tumor mass

palpable after ~3-4 weeks (Fig.5B). This finding was similar to a prior study on using a

comparable histotripsy system to treat hepatocellular carcinoma, where 2/6 of their

treated mice saw tumor size completely deplete before regrowing in

immunocompromised mice [17].

Page 119: Determining the Oncological and Immunological Effects of

82

Overall, the results of this study continue to support our hypothesis that

histotripsy has the potential to be a viable treatment method for CC, representing the

first completely non-invasive and non-thermal ablation method for this application.

However, results also demonstrate limitations with the animal models used in this study.

For instance, looking to the future, with how slowly these PDX tumors grew (Fig.5), the

lack of metastasis (Fig.7), and the histological features that are similar to human tumors

[45], this model would be useful for studying the possibility of using histotripsy to non-

invasively manage disease through multiple treatments of recurring tumors. Additionally,

in this study we were limited to a safety margin that prevented aggressive margins that

would have the potential to completely eliminate the tumors. When translating this

consideration to larger animal pre-clinical work or human trials, it should be noted that

these off-target damages have been established in the murine model. Future studies

would be needed to determine the dose safety between tumors and margin, critical

structures, such as the intestines. While this study suggests that a complete ablation

may be limited due to safety concerns, overall, these results suggest that histotripsy

would be a usefully palliative option for patients that do not qualify for curative surgeries.

3.7 Conclusion

This study demonstrates the potential to utilize histotripsy in the treatment of CC,

with results showing a maximum average tumor diameter reduction of 73% in a

subcutaneous CC model after 26 days. There were no significant side effects using the

high dose and conservative margins (n=6), with 3 of 6 treated tumors reduced in

Page 120: Determining the Oncological and Immunological Effects of

83

diameter to an undetectable size in 2.5 weeks of treatment as the ablation homogenate

was reabsorbed. The treated animals were found to have significantly increased

progression-free and overall survival, and no metastases were observed in any animals.

Additionally, due to the subcutaneous placement of the tumors in this study, we were

limited to a safety margin that prevented aggressive, positive margins that would have

the potential to completely eliminate the tumors in all animals. Overall, the results of this

study support the hypothesis that histotripsy has the potential to be a viable treatment

method for CC, representing the first completely non-invasive and non-thermal ablation

method for this application.

3.8 Acknowledgment

This research was supported by a grant from the Focused Ultrasound

Foundation (FUSF-RAP-S-18-00011) and was conducted in memory of Andrew J.

Lockhart, who passed away from cholangiocarcinoma on September 30, 2016. The

authors encourage anyone interested to consider a donation to the Focused Ultrasound

Foundation in Andrew’s name.

The authors would also like to thank the ICTAS Center for Engineered Health, the

Virginia Tech Department of Biomedical Engineering and Mechanics, Virginia Tech

Carilion School of Medicine, and the Virginia-Maryland College of Veterinary Medicine

for their support of this study.

Page 121: Determining the Oncological and Immunological Effects of

84

3.9 References

[1] ACS, "Cancer Facts & Figures 2020," American Cancer Society, 2020.

[2] E. U. Cidon, "Resectable cholangiocarcinoma: reviewing the role of adjuvant

strategies," Clinical Medicine Insights: Oncology, vol. 10, p. CMO. S32821, 2016.

[3] M. Jansen, R. Van Hillegersberg, R. Chamuleau, O. Van Delden, D. Gouma, and

T. Van Gulik, "Outcome of regional and local ablative therapies for hepatocellular

carcinoma: a collective review," European Journal of Surgical Oncology (EJSO),

vol. 31, no. 4, pp. 331-347, 2005.

[4] L. Rossi et al., "Current approach in the treatment of hepatocellular carcinoma,"

World journal of gastrointestinal oncology, vol. 2, no. 9, p. 348, 2010.

[5] S. K. Venkatesh et al., "MR elastography of liver tumors: preliminary results,"

American Journal of Roentgenology, vol. 190, no. 6, pp. 1534-1540, 2008.

[6] D. M. Nagorney, J. H. Donohue, M. B. Farnell, C. D. Schleck, and D. M. Ilstrup,

"Outcomes after curative resections of cholangiocarcinoma," Archives of surgery,

vol. 128, no. 8, pp. 871-878, 1993.

[7] K. Feng et al., "A randomized controlled trial of radiofrequency ablation and

surgical resection in the treatment of small hepatocellular carcinoma," Journal of

hepatology, vol. 57, no. 4, pp. 794-802, Oct 2012, doi:

10.1016/j.jhep.2012.05.007.

[8] M. S. Chen et al., "A prospective randomized trial comparing percutaneous local

ablative therapy and partial hepatectomy for small hepatocellular carcinoma," (in

Page 122: Determining the Oncological and Immunological Effects of

85

eng), Ann Surg, Randomized Controlled Trial, Research Support, Non-U.S. Gov't

vol. 243, no. 3, pp. 321-8, Mar 2006, doi: 10.1097/01.sla.0000201480.65519.b8.

[9] R. Salem et al., "Institutional Decision to Adopt Y90 as Primary Treatment for

HCC Informed by a 1,000-patient 15-year Experience," Hepatology, Dec 1 2017,

doi: 10.1002/hep.29691.

[10] A. Forner, J. M. Llovet, and J. Bruix, "Hepatocellular carcinoma," Lancet, vol.

379, no. 9822, pp. 1245-55, Mar 31 2012, doi: 10.1016/S0140-6736(11)61347-0.

[11] L. European Association for the Study of the, R. European Organisation for, and

C. Treatment of, "EASL–EORTC Clinical Practice Guidelines: Management of

hepatocellular carcinoma," Journal of hepatology, vol. 56, no. 4, pp. 908-943,

2012. [Online]. Available:

http://linkinghub.elsevier.com/retrieve/pii/S0168827811008737?showall=true.

[12] A. Gillams et al., "Thermal ablation of colorectal liver metastases: a position

paper by an international panel of ablation experts, The Interventional Oncology

Sans Frontieres meeting 2013," Eur Radiol, vol. 25, no. 12, pp. 3438-54, Dec

2015, doi: 10.1007/s00330-015-3779-z.

[13] K. Han, H. K. Ko, K. W. Kim, H. J. Won, Y. M. Shin, and P. N. Kim,

"Radiofrequency ablation in the treatment of unresectable intrahepatic

cholangiocarcinoma: systematic review and meta-analysis," Journal of Vascular

and Interventional Radiology, vol. 26, no. 7, pp. 943-948, 2015.

[14] E. A. Takahashi et al., "Thermal ablation of intrahepatic cholangiocarcinoma:

safety, efficacy, and factors affecting local tumor progression," Abdominal

Radiology, vol. 43, no. 12, pp. 3487-3492, 2018.

Page 123: Determining the Oncological and Immunological Effects of

86

[15] E. Vlaisavljevich et al., "Non-Invasive Ultrasound Liver Ablation Using Histotripsy:

Chronic Study in an In Vivo Rodent Model," Ultrasound Med Biol, vol. 42, no. 8,

pp. 1890-902, Aug 2016, doi: 10.1016/j.ultrasmedbio.2016.03.018.

[16] E. Vlaisavljevich et al., "Image-guided non-invasive ultrasound liver ablation

using histotripsy: Feasibility study in an in vivo porcine model," Ultrasound in

medicine & biology, vol. 39, no. 8, pp. 1398-1409, 2013.

[17] T. Worlikar et al., "Histotripsy for Non-Invasive Ablation of Hepatocellular

Carcinoma (HCC) Tumor in a Subcutaneous Xenograft Murine Model," in 2018

40th Annual International Conference of the IEEE Engineering in Medicine and

Biology Society (EMBC), 2018: IEEE, pp. 6064-6067.

[18] A. D. Maxwell, C. A. Cain, T. L. Hall, J. B. Fowlkes, and Z. Xu, "Probability of

cavitation for single ultrasound pulses applied to tissues and tissue-mimicking

materials," Ultrasound in medicine & biology, vol. 39, no. 3, pp. 449-465, 2013.

[19] E. Vlaisavljevich et al., "Effects of Ultrasound Frequency and Tissue Stiffness on

the Histotripsy Intrinsic Threshold for Cavitation," Ultrasound Med Biol, 2015.

[20] Z. Xu et al., "Controlled ultrasound tissue erosion," IEEE transactions on

ultrasonics, ferroelectrics, and frequency control, vol. 51, no. 6, pp. 726-736,

2004.

[21] Z. Xu, M. Raghavan, T. L. Hall, M.-A. Mycek, J. B. Fowlkes, and C. A. Cain,

"Evolution of bubble clouds produced in pulsed cavitational ultrasound therapy -

histotripsy," IEEE Trans Ultrason Ferroelectr Freq Control., vol. 55, no. 5, pp.

1122-1132, 2008.

Page 124: Determining the Oncological and Immunological Effects of

87

[22] E. Vlaisavljevich, A. Maxwell, M. Warnez, E. Johnsen, C. Cain, and Z. Xu,

"Histotripsy-induced cavitation cloud initiation thresholds in tissues of different

mechanical properties," (in eng), IEEE Transactions on Ultrasonics,

Ferroelectrics, and Frequency Control, vol. 61, no. 2, pp. 341-52, Feb 2014, doi:

10.1109/TUFFC.2014.6722618.

[23] E. Vlaisavljevich, A. Maxwell, L. Mancia, E. Johnsen, C. Cain, and Z. Xu,

"Visualizing the Histotripsy Process: Bubble Cloud-Cancer Cell Interactions in a

Tissue-Mimicking Environment," (in Eng), Ultrasound Med Biol, Jul 8 2016, doi:

S0301-5629(16)30099-0 [pii] 10.1016/j.ultrasmedbio.2016.05.018.

[24] K. C. Longo et al., "Histotripsy Ablations in a Porcine Liver Model: Feasibility of

Respiratory Motion Compensation by Alteration of the Ablation Zone Prescription

Shape," CardioVascular and Interventional Radiology, pp. 1-7, 2020.

[25] T. Worlikar et al., "Non-Invasive Orthotopic Liver Tumor Ablation Using

Histotripsy in an in Vivo Rodent Hepatocellular Carcinoma (HCC) Model,"

presented at the Society of Interventional Oncology, Boston, MA, USA, 2019.

[26] K. C. Longo et al., "Robotically Assisted Sonic Therapy (RAST) for Noninvasive

Hepatic Ablation in a Porcine Model: Mitigation of Body Wall Damage with a

Modified Pulse Sequence," Cardiovasc Intervent Radiol, vol. 42, no. 7, pp. 1016-

1023, Jul 2019, doi: 10.1007/s00270-019-02215-8.

[27] E. Vlaisavljevich et al., "Non-invasive liver ablation using histotripsy: preclinical

safety study in an in vivo porcine model," Ultrasound in medicine & biology, vol.

43, no. 6, pp. 1237-1251, 2017.

Page 125: Determining the Oncological and Immunological Effects of

88

[28] T. Worlikar et al., "Non-invasive liver cancer ablation using histotripsy in an in

vivo murine hepatocellular carcinoma (HCC) model," in Ultrasonics Symposium

(IUS), 2017 IEEE International, 2017: IEEE, pp. 1-1.

[29] J. Vidal Jove et al., "Phase I Study of Safety and Efficacy of Hepatic Histotripsy:

Preliminary Results of First in Man Experience with Robotically-Assisted Sonic

Therapy," presented at the International Society of Therapeutic Ultrasound,

Barcelona, Spain, 2019.

[30] E. Vlaisavljevich, Y. Kim, G. Owens, W. Roberts, C. Cain, and Z. Xu, "Effects of

tissue mechanical properties on susceptibility to histotripsy-induced tissue

damage," Physics in Medicine & Biology, vol. 59, no. 2, p. 253, 2013.

[31] E. Vlaisavljevich, Z. Xu, A. Arvidson, L. Jin, W. Roberts, and C. Cain, "Effects of

thermal preconditioning on tissue susceptibility to histotripsy," Ultrasound in

medicine & biology, vol. 41, no. 11, pp. 2938-2954, 2015.

[32] T. L. Hall, C. R. Hempel, K. Wojno, Z. Xu, C. A. Cain, and W. W. Roberts,

"Histotripsy of the prostate: dose effects in a chronic canine model," Urology, vol.

74, no. 4, pp. 932-937, 2009.

[33] A. Lake, Z. Xu, J. Wilkinson, C. Cain, and W. Roberts, "Renal ablation by

histotripsy—does it spare the collecting system?," The Journal of urology, vol.

179, no. 3, pp. 1150-1154, 2008.

[34] C. R. Hempel, T. L. Hall, C. A. Cain, J. B. Fowlkes, Z. Xu, and W. W. Roberts,

"Histotripsy fractionation of prostate tissue: local effects and systemic response

in a canine model," The Journal of urology, vol. 185, no. 4, pp. 1484-1489, 2011.

Page 126: Determining the Oncological and Immunological Effects of

89

[35] E. Vlaisavljevich, Y. Kim, G. Owens, W. Roberts, C. Cain, and Z. Xu, "Effects of

tissue mechanical properties on susceptibility to histotripsy-induced tissue

damage," (in eng), Phys Med Biol, vol. 59, no. 2, pp. 253-70, Jan 20 2014, doi:

10.1088/0031-9155/59/2/253.

[36] V. A. Khokhlova et al., "Histotripsy methods in mechanical disintegration of

tissue: Towards clinical applications," International journal of hyperthermia, vol.

31, no. 2, pp. 145-162, 2015.

[37] A. Hendricks et al., "Histotripsy for the Treatment of Cholangiocarcinoma Liver

Tumors: In Vivo Feasibility and Ex Vivo Dosimetry Study," IEEE Transactions on

Ultrasonics, Ferroelectrics, and Frequency Control, 2021.

[38] A. Guibal et al., "Evaluation of shearwave elastography for the characterisation of

focal liver lesions on ultrasound," European radiology, vol. 23, no. 4, pp. 1138-

1149, 2013.

[39] R. M. Brock et al., "Patient derived xenografts expand human primary pancreatic

tumor tissue availability for ex vivo irreversible electroporation testing," Frontiers

in Oncology, vol. 10, 2020.

[40] A. Hendricks et al., "Histotripsy initiates local and systemic immunological

response and reduces tumor burden in breast cancer

AD Hendricks, J Howell, R Schmieley, S Kozlov, A Simon, ..." The Journal of

Immunology, vol. 202, 2019.

[41] A. D. Hendricks et al., "Determining the mechanism of the immune response to

histotripsy ablation of pancreatic cancer," ed: Am Assoc Immnol, 2020.

Page 127: Determining the Oncological and Immunological Effects of

90

[42] S. Qu et al., "Non-thermal histotripsy tumor ablation promotes abscopal immune

responses that enhance cancer immunotherapy," Journal for ImmunoTherapy of

Cancer, vol. 8, no. 1, 2020.

[43] A. Hendricks-Wenger et al., "Histotripsy Ablation Alters the Tumor

Microenvironment and Promotes Immune System Activation in a Subcutaneous

Model of Pancreatic Cancer," IEEE Transactions on Ultrasonics, Ferroelectrics,

and Frequency Control, 2021.

[44] L. E. Dobrolecki et al., "Patient-derived xenograft (PDX) models in basic and

translational breast cancer research," Cancer and Metastasis Reviews, vol. 35,

no. 4, pp. 547-573, 2016.

[45] B. Blechacz, M. Komuta, T. Roskams, and G. J. Gores, "Clinical diagnosis and

staging of cholangiocarcinoma," Nature reviews Gastroenterology & hepatology,

vol. 8, no. 9, pp. 512-522, 2011.

Page 128: Determining the Oncological and Immunological Effects of

91

Chapter 4. Histotripsy Ablation in Preclinical Animal Models of Cancer and Spontaneous Tumors in Veterinary Patients

Alissa Hendricks-Wenger, Lauren Arnold, Jessica Gannon, Alex Simon, Neha Singh, Hannah Sheppard, Margaret A. Nagai-Singer, Khan Mohammad Imran1, Kiho Lee,

Sherrie Clark-Deener, Christopher Byron, Martha M. Larson, John H. Rossmeisl, Sheryl L. Coutermarsh-Ott, Nikolaos Dervisis, Shawna Klahn, Joanne Tuohy, Irving C. Allen,

Eli Vlaisavljevich This chapter is excerpted from a manuscript submitted to IEEE Transactions in UFFC.

Page 129: Determining the Oncological and Immunological Effects of

92

4.1 Abstract

New therapeutic strategies are direly needed in the fight against cancer. Over the last

decade, several focal tumor ablation strategies have emerged as stand-alone or

combination therapies. Histotripsy, an ultrasound ablation method that destroys tissue

through the precise control of acoustic cavitation, has emerged as the first completely

non-invasive, non-thermal, and non-ionizing ablation method for the treatment of

cancer. Histotripsy does not share the limitations of thermal ablation, can produce

consistent and rapid ablations, and even near critical structures. Additional benefits

include real-time image-guidance, high precision, and the ability to treat tumors of any

predetermined size and shape. Unfortunately, the lack of clinically and physiologically

relevant pre-clinical cancer models is often a significant limitation with all focal tumor

ablation strategies. The majority of studies testing histotripsy for cancer treatment have

focused on small animal models, which have been critical in moving this field forward

and will continue to be essential for providing mechanistic insight. However, these

models have significant limitations in terms of scale and translational value. To address

these limitations, a diverse range of large animal models and spontaneous tumor

studies in veterinary patients have emerged to complement existing rodent models.

These models and veterinary patients have proven to be excellent at providing realistic

models for developing and testing histotripsy devices and techniques designed for

future use in human patients. Here, we compare histotripsy ablation across a

representative spectrum of preclinical animal models and spontaneous tumors in

Page 130: Determining the Oncological and Immunological Effects of

93

veterinary patients. Together, these studies demonstrate the utility and advantages of

these bench-to-kennel-to-bedside approaches in the development of histotripsy.

Page 131: Determining the Oncological and Immunological Effects of

94

4.2 Introduction

Histotripsy is a focused ultrasound ablation method that destroys tissue through the

precise control of acoustic cavitation [1-4]. Using microsecond long, high-pressure

pulses applied by a focused ultrasound transducer coupled to the patient, a histotripsy

bubble cloud is generated non-invasively inside the targeted tissue [5-9]. The rapid

expansion and collapse of these bubbles result in complete disruption of the target

tissue into acellular debris [10]. As a non-thermal ablation method, histotripsy has

shown the potential to overcome the limitations associated with thermal ablation. For

instance, histotripsy has been shown to produce consistent and complete ablation in the

liver, which is highly vascular, when applied through abdominal or transcostal acoustic

windows [1, 3, 11, 12]. Histotripsy has also shown the ability to ablate tissue near vital

structures such as major vessels, bile ducts, and nerves while preserving these

structures [1, 3, 9, 13, 14]. Due to these features, histotripsy is currently being

developed for multiple clinical applications, most notably for the treatment of cancer [4,

15-17]. However, despite the promise of histotripsy and the many positive results

reported in preclinical studies, to date, only a single human clinical trial has been

conducted to test histotripsy for the treatment of cancer [18]. This recent Phase I clinical

trial was conducted in non-curative patients with multifocal primary and metastatic liver

tumors, with results showing histotripsy could safely target and ablate targeted liver

tumors [18]. To our knowledge, no other human clinical trials of histotripsy for the

Page 132: Determining the Oncological and Immunological Effects of

95

treatment of cancer have been conducted, limiting the potential widespread translation

of histotripsy into clinical practice as an improved non-invasive tumor ablation method.

Similar to other focal tumor ablation modalities, one of the primary factors limiting the

rapid translation of histotripsy into the clinic has been the lack of appropriate animal

models for studying the safety, effectiveness, and optimization of histotripsy for the

treatment of different cancers. Previous efforts to develop histotripsy for specific

oncology applications have been split into separate studies testing human-scale

histotripsy systems for targeting and ablating healthy tissue in large animal models that

have similar anatomy and physiology to human patients (primarily pigs) [1, 3, 11, 12].

These studies are typically complemented by separately testing histotripsy tumor

ablation in rodents (mice, rats) using customized small animal devices that allow

histotripsy to be applied in this setting, albeit not using the systems or acoustic

parameters that will ultimately be developed for use in humans [2, 4, 19, 20].

Murine models are the dominant in vivo model in the biomedical engineering and

cancer biology fields and are essential in acquiring fundamental data. Mice are easy to

house and handle, relatively inexpensive, share many genetic similarities to humans,

and there are many transgenic and knock-out lines available to study specific disease

mechanisms, proteins, and pathways [21-26]. In typical mouse models used to study

tumor ablation modalities, the vast majority of studies are mouse or human cell line

based. While these models offer excellent reproducibility and standardization, they

rarely recapitulate the clinical, in situ, microenvironment found in patients or critical

biological hallmarks of the original tumor of origin. This is especially true in

subcutaneous, flank generated tumors. Orthotopic tumors that are transplanted into the

Page 133: Determining the Oncological and Immunological Effects of

96

organ of interest can provide improved insight but still suffer from cell line specific

limitations related to cancer biology. Spontaneous and induced tumors in small animals

are more physiologically relevant to human patients. However, the inability to predict

tumor development, location, progression, and lack of reproducibility are all limitations

to these models. Likewise, the lack of translational acoustic windows, limited imaging,

lack of precision, and lack of physiological relevance are all major limitations of small

animal models in the development of histotripsy and other focused ultrasound

modalities. While critical for proof-of-concept testing and mechanistic insight, the

miniaturization of the model has significantly slowed the rate of clinical translation and

made it difficult to reliably test the effectiveness of clinical prototype histotripsy systems

for imaging, targeting, and effectively treating different tumors types. As a result, there is

a significant need for improved preclinical large animal tumor models for studying

histotripsy tumor ablation using clinically relevant devices and treatment parameters to

enable the more rapid and successful translation of histotripsy into clinical practice for

the treatment of cancer in humans.

Large animal models can address many of the shortcomings often cited for

rodents, in many cases offering direct translation from animal to human. For example,

the same tumor ablation systems can typically be utilized, similar clinical/surgical

techniques can be refined, and many large animal models accurately recapitulate

human anatomical and physiological features. This is critical for imaging and

ultrasound-based modalities such as histotripsy. However, the increased size and

complexity of the species results in several significant limitations typically associated

with cost, the requirement for specialized housing, and the need for unique institutional

Page 134: Determining the Oncological and Immunological Effects of

97

resources. Most large animal models are based on spontaneous tumor development,

which has some of the same limitations commonly cited for small animal studies.

Recent technological advancements in gene editing technologies have largely negated

these issues, specifically in pig models. In general, pigs have yet to play a significant

role in experimental oncology [27]. However, with the rise of technologies capable of

inducing genetic modifications in large mammals, such as CRISPRs and TALONs, there

is a renewed interest in the pig as a cancer model. This is especially true in the area of

medical devices, where the utilization of the same devices, instruments, and clinical

practice can be utilized in pigs and humans, enabling a more rapid translation to clinical

trials. Over the last decade, genetically modified porcine cancer models have been

generated targeting APC, TP53, KRAS, GLI2, V-H-RAS, and BRACA1[27]. Indeed, the

recently developed “Oncopig” cancer model (OCM) has the potential to bring the pig to

the forefront of tumor models [28]. These novel transgenic swine models recapitulate

human cancer through Cre recombinase induced site and cell-type specific expression

of KRAS and TP53 transgenes [29]. To express Cre, pigs must be either crossed with

transgenic animals expressing cell type specific Cre or direct injection into the tissue

type of interest with an adenoviral vector encoding Cre recombinase [28]. Both KRAS

and TP53 mutations are common in multiple cancers and multiple OCM models appear

to be currently under development [28].

The OCM model is expected to have several strengths, including an accurate

recapitulation of the tumor microenvironment, accurate immune system profile, and the

outbred animals are expected to better model human patients [28]. However, the

reliance on Cre recombinase and associated delivery vectors will result in an

Page 135: Determining the Oncological and Immunological Effects of

98

unpredictable number of tumors that form in random locations. Likewise, while tumor

development in the OCM model is rapid, it is highly difficult to predict tumor progression

in this model. This can add significant cost and time. As an alternative strategy,

research teams have focused on immunocompromised pig models, where deletions in

the RAG2 and IL2RG genes have generated animals with similar characteristics to NOD

scid gamma (NSG) mice, which are susceptible to human tissue engraftment [30].

Similar to NSG mice, human tumor cell lines, and patient-derived xenograft (PDX)

tissue can be engrafted. This allows a highly predictable tumor that can be surgically

placed subcutaneously or orthotopically in a human-sized animal with similar anatomy

and physiology. Tumors can be expanded to increase tissue availability for ex vivo

studies or surgically placed near critical structural elements (such as blood vessels or

nerve bundles) to evaluate and model tumor ablation in hard to treat locations within the

organ. Another advantage of this strategy is the use of human cells in a pig model. This

allows robust identification, evaluation, and quantification of the ablation zone and

metastatic tumors at the histopathologic level using human antibodies in porcine

tissues. The lack of a functional immune system is a clear disadvantage of

immunocompromised pig models as they require housing under highly specialized

germfree conditions that are only available at a limited number of institutions.

Beyond tumor models, spontaneous cancers in pet animals offer unique

translational research opportunities for cancer imaging and device development that

provide a robust combination of clinical and physiological relevance to humans. Indeed,

comparative oncology studies have revealed significant similarities between many

canine and human cancers [31, 32]. Companion animals, like humans, are genetically

Page 136: Determining the Oncological and Immunological Effects of

99

diverse and are exposed to many of the same environmental influences as their owners.

Canine cancer patients have been successfully used in numerous studies to provide

translatable pharmacokinetic and pharmacodynamic data [33-38], and these trials have

been critical for determining the scheduling/dosing of small molecular inhibitors in

humans [39]. The spontaneous nature of canine cancers, their biologic similarities to

human cancers, and the anatomic similarities between dogs and people offer highly

relevant clinical conditions for device development.

In this work, we provide an overview of small animal tumor models currently being

used for histotripsy studies, as well as, emerging large animal models under

development by our group for studying histotripsy tumor ablation. In the first part of this

work, we provide a comparison of the histotripsy experimental set-ups commonly used

in small and large animal experiments. We then show representative results of

histotripsy tumor ablation in small animal (mice) cancer models including examples of

subcutaneous, orthotopic, and patient-derived xenograft (PDX) models. Next, we show

results from a recently developed immunocompromised RAG2/IL2RG deficient pig

tumor model that better mimic human patient anatomy and physiology compared to the

mouse models. Finally, we discuss the opportunity to investigate the safety, efficacy,

and optimization of histotripsy in veterinary populations of dogs with spontaneous

tumors and provide an overview of the canine oncology patients at our location as well

as examples of our preliminary studies of histotripsy for the treatment of a variety of

cancers in these veterinary patients. Overall, the results of this study will provide an

overview of the benefits and limitations of currently available animal models and

highlight the potential to utilize emerging large animal tumor models and veterinary

Page 137: Determining the Oncological and Immunological Effects of

100

cancer patients as a means to enable more rapid and reliable clinical translation of

histotripsy and other emerging FUS methods for the treatment of patients with a wide

range of cancers.

4.3 Procedures

4.3.1 Histotripsy Systems and Pressure Calibrations

Two separate histotripsy systems were used in this study for the small animal and

large animal in vivo histotripsy studies. The small animal in vivo experiments were

conducted using a custom 8-element 1MHz histotripsy transducer with a geometric

focus of 36 mm, an aperture size of 52.7 mm, and an f-number of 0.68 (Fig.1B). The

transducer was driven via a custom high-voltage pulser designed to generate short

therapy pulses of <2 cycles controlled by a field-programmable gate array (FPGA)

board (Altera DE0-Nano Terasic Technology, Dover, DE, USA) programmed for

histotripsy therapy pulsing. The transducer was positioned in a tank of degassed water

beneath a custom-designed mouse surgical stage and attached to a computer-guided

3-D positioning system with 0.05 mm motor resolution to control the automated

volumetric treatments (Fig.1A). A linear ultrasound imaging probe with a frequency

range of 10-18 MHz (L18-10L30H-4, Telemed, Lithuania, EU) was coaxially aligned

inside the transducer for treatment guidance and monitoring (Fig.1C). The transducer

was powered by a high voltage DC power supply (GENH750W, TDK-Lambda), and the

system was controlled using a custom user-interface operated through MATLAB

Page 138: Determining the Oncological and Immunological Effects of

101

(MathWorks). This transducer design has been used previously for multiple small

animal histotripsy studies in mouse and rat models [2, 4, 19, 20].

Figure 4-1 Example Small Animal Histotripsy System. (A) A small animal system consisting of a 1) 1 MHz therapy transducer with an 2) imaging transducer coaxially aligned and mounted to a 3) 3D motorized positioning system to deliver hisotripsy to target submerged in degassed water on a 5) subject stage. A custom-built electronic driving system is controlled with computer software. (B) Linear ultrasound imaging probe coaxially aligned with 1 MHz therapy transducer used to (C) treat tumors in-vivo. (D) Acoustic waveform for 1 MHz transducer collected with a hydrophone at a peak negative pressure of ~10 MPa. (E) 1 MHz bubble cloud captured on a high-speed camera.

For the large animal in vivo experiments, histotripsy was applied using a custom 32-

element 500 kHz array transducer with a geometric focus of 78 mm, an elevational

aperture size of 112 mm, and a transverse aperture size of 128 mm, with corresponding

f-numbers of 0.61 (transverse) and 0.70 (elevational) (Fig.2B). The transducer was

driven via custom high-voltage pulser designed to generate short therapy pulses of <2

cycles controlled by a field-programmable gate array (FPGA) board (Altera DE0-Nano

Terasic Technology, Dover, DE, USA) programmed for histotripsy therapy pulsing. The

transducer was fixed onto a prototype clinical histotripsy treatment cart (HistoSonics,

Ann Arbor, MI, USA) that included an ultrasound imaging system, robotic micro-

positioner, and customized for applying volumetric histotripsy ablations (Fig.2A). A 3

Page 139: Determining the Oncological and Immunological Effects of

102

MHz curvilinear imaging probe (Model C5-2, Analogic Corp., Peabody, MA) was

coaxially aligned through a central hole in the therapy transducer in order to allow real-

time treatment guidance and monitoring (Fig.2B). The transducer was powered by a

high voltage DC power supply (GENH750W, TDK-Lambda) and the transducer was

controlled using a custom user-interface operated through MATLAB (MathWorks) which

was triggered from the HistoSonics system. This overall system is similar to the one

used in recent large animal histotripsy studies conducted on healthy pigs as well as a

recent Phase I clinical trial of histotripsy for treating liver cancer patients [1, 18], with the

only difference being the custom 32-element transducer that was used in this study in

place of the clinical histotripsy transducer used in the prior work.

Figure 4-2 Example Large Animal Histotripsy System. (A) Histotripsy system designed for liver cancer ablation consisting of 1) therapy and 2) imaging transducers attached to an 3) articulating arm with a 4) motorized micro-positioner controlled by 5) custom software. (B) Image of therapy transducer with co-axially aligned imaging probe used for (C) previous in vivo liver studies. (D) Acoustic waveform for 500 kHz transducer collected with a hydrophone at a peak negative pressure of ~10 MPa. (E) 500 kHz bubble captured on a high-speed camera.

Page 140: Determining the Oncological and Immunological Effects of

103

4.3.2 Hydrophone Focal Pressure Measurements, Beam Profiles, and Optical Imaging

Focal pressure waveforms for the 1MHz and 500kHz transducers were measured by

a high sensitivity rod hydrophone (HNR-0500, Onda Corporation, Sunnyvale, CA, USA)

and a custom-built fiber optic probe hydrophone (FOPH) [40, 41] in degassed water at

the focal point of each transducer. The rod hydrophone was used to measure the 1D

focal beam profiles of the transducer in the lateral, elevational, and axial directions at a

peak negative pressure (p-) of ~1.8 MPa. Focal pressures were directly measured with

the FOPH up to a p- of ~20 MPa. Pressure waveforms at higher p- were estimated by a

linear extrapolation [42] since p- values above ~20MPa couldn’t be directly measured

due to cavitation on the tip of the FOPH fiber. All waveforms were measured using a

Tektronix TBS2000 series oscilloscope at a sample rate of 500MS/s, with the

waveforms averaged over 128 pulses and recorded in MATLAB.

The bubble clouds generated by each transducer were characterized using high-

speed optical imaging as described in previous studies [43, 44]. Briefly, the focal region

inside 1% agarose tissue phantoms was imaged after acoustic propagation for each

pulse using the 1 MHz and 500 kHz therapy transducers described above. Optical

imaging was performed using a high-speed camera (FLIR Blackfly S monochrome,

BFS-U3-32S4M-C 3.2 MP, 118 FPS, Sony IMX252, Mono, FLIR Integrated Imaging

Solutions, Richmond, BC, Canada) and a 100 mm F2.8 Macro lens (Tokina AT-X Pro,

Kenko Tokina Co., LTD, Tokyo, Japan). The camera was externally triggered to record

one image per applied pulse, and the tissue phantom was backlit by a custom-built

pulsed white-light LED strobe light capable of high-speed triggering with 1 μs

Page 141: Determining the Oncological and Immunological Effects of

104

exposures. Strobe duration was kept low (3-5 µs) and all exposures were centered at a

delay of 8.5 µs after pulse arrival at the focus. This timing schema was previously

demonstrated as the optimum delay for visualizing complete bubble clouds [43, 44]. The

bubble cloud dimensions observed on optical imaging were subsequently used to

determine the spacing between adjacent treatment points in in vivo tumor ablation

experiments.

4.3.3 Murine Tumor Models

The ability of histotripsy to ablate tumors in in vivo mouse models was tested using

the 1 MHz small animal histotripsy system to target tumors in three murine cancer

models including 1) a subcutaneous renal cell carcinoma (RCC) model, 2) a patient-

derived xenograft (PDX) subcutaneous cholangiocarcimona model, and 3) a 4T1

orthotopic mammary tumor model. All procedures in this study were conducted in

accordance with the ARRIVE guidelines and approved by the IACUC at Virginia Tech.

In the subcutaneous RCC model, male BALB/cCr mice (n=4) were injected in the right

flank with 6x106 Renca RCC cells (ATCC CRL-2947) in Matrigel. In the subcutaneous

cholangiocarcinoma (CC) PDX model, female Nod scid gamma (NSG) mice (n=4) with

subcutaneous flank PDX CC tumors were commercially acquired from the Jackson

Laboratory (TM01225), through a process previously described [45]. In the orthotopic

mammary tumor model, female BALB/C mice (n=4) were injected with 1.2 X 106 murine

4T1 mammary tumor cells (ATCC CRL-2539) in the abdominal mammary glands. Once

tumors were palpable, animals were monitored three times weekly and tumor diameter

Page 142: Determining the Oncological and Immunological Effects of

105

was determined by taking two perpendicular measurements, one of which was the

widest point of the tumor. A single tumor diameter was calculated as the square root of

the product of the two measurements. The mice underwent histotripsy treatment when

their tumors reached the targeted treatment size ~0.5-1.0 cm in diameter.

For all tumors, histotripsy was applied after each tumor reached the

predetermined target size. For all treatments, mice were anesthetized with inhaled

isoflurane and secured to a platform over a tank of degassed water, with the

temperature maintained between 35-39°C. Before treatment, fur was removed over the

tumor and the surrounding area by applying Nair (Naircare, Ewing, NJ, USA) for 1-2

minutes and removed by wiping off with a wet paper towel. For treatment, mice were

anesthetized with isoflurane (1.5 L/min oxygen flow rate with 1.0-2.5% isoflurane) and

placed prone on the subject stage with their tumor submerged underwater (Fig.1A).

Animals’ respiratory rate and rhythm were monitored during treatment by trained

personnel. Histotripsy was then applied to a predetermined volume of the tumor using

the custom 1MHz small animal transducer described above and shown in Figure 1.

During treatment, histotripsy was applied using single cycle pulses applied at a pulse

repetition frequency (PRF) of 250 Hz with a 1 second dwell time at each treatment

point. To apply a volumetric treatment, the focus was raster-scanned through a series of

treatment points spaced by 1.5 mm in the axial direction and 0.5 in the lateral for the

Renca and 4T1 tumors. Given that the CC tumors are stiffer and more resistant to

histotripsy, a PRF of 500 Hz with a 1 second dwell time was used to double the per

point dose and the spacing was reduced to 0.75 mm and 0.25 mm to increase the

overlap between adjacent treatment points.

Page 143: Determining the Oncological and Immunological Effects of

106

4.3.4 RAG2/IL2RG Deficient Porcine Tumor Model

An ideal animal model for studying histotripsy tumor ablation using clinical prototype

systems would 1) be implantable so that specific locations could be chosen for the

hypothesis being tested, 2) allow implantation of human tumor cell lines to closely mimic

the natural history and composition of human tumors, and 3) be in an animal large

enough to evaluate human-scale devices. To accomplish these goals, our team has

recently developed RAG2/IL2RG double knockout pigs [46-52]. Oocytes were aspirated

from sow ovaries and matured in maturation medium (TCM-199 medium supplemented

with 0.5 IU ml−1 FSH, 0.5 IU ml−1 LH, 0.82 mM cysteine, 3.02 mM glucose, 0.91 mM

sodium pyruvate and 10 ng ml−1 EGF) for 40 – 44 hours. After in vitro maturation,

cumulus cells were removed by vortexing in the presence of hyaluronidase, then mature

oocytes extruding the first polar body were transferred to fertilization media (modified

Tris-buffered medium supplemented with 113.1 mM NaCl, 3 mM KCl, 7.5 mM CaCl2,

11 mM glucose, 20 mM Tris, 2 mM caffeine, 5 mM sodium pyruvate and 2 mg ml−1

BSA). Extended semen was washed with PBS, then introduced into the IVF dishes

containing oocytes. The gametes were co-incubated for 5 hours at 38.5 °C and 5%

CO2. Microinjection was conducted in manipulation media (TCM199 with 0.6 mM

NaHCO3, 2.9 mM HEPES, 30 mM NaCl, 10 ng ml−1 gentamicin and 3 mg ml−1 BSA)

covered with mineral oil on the heated stage of a Nikon inverted microscope (Nikon

Corporation, Tokyo, Japan). Two sgRNAs targeting the RAG2 gene, three sgRNAs

targeting the IL2RG gene, and Cas9 mRNA (10 ng/µl sgRNA each and 20 ng/μl Cas9

mRNA) were introduced into the cytoplasm of the presumable zygotes using a FemtoJet

Page 144: Determining the Oncological and Immunological Effects of

107

microinjector (Eppendorf, Hamburg, Germany). The injected embryos were moved to

culture media supplemented with 10 ng ml−1 GM-CSF and incubated at 38.5 °C, 5%

CO2, and 5% O2 until embryo transfer [53]. The cultured embryos were transferred into

a surrogate gilt at day 4 post-IVF by surgically transferring into the oviduct. Pregnancy

was determined by ultrasound at around day 30 of gestation. To genotype the mutations

generated by CRISPR/Cas9 system, genomic DNA was isolated from tail-tips of the

newborn piglets using PureLink Genomic DNA kit (Thermo Fisher Scientific) following

the manufacturer’s instructions. Primers flanking projected double-strand break (DSB)

sites were designed to amplify the target region of RAG2 and IL2RG genes. The target

regions were PCR amplified using Dream Taq DNA Polymerase (Thermo Fisher

Scientific). PCR conditions were as follows: initial denature at 95 °C for 2 min; denature

at 95 °C for 30 sec, annealing at 60 °C for 30 sec, and extension at 72 °C for 30 sec for

34 cycles; 72 °C for 5 min; and holding at 4 °C. The PCR amplicons were sequenced to

determine the mutation types generated by the CRISPR/Cas9 system.

Immunocompromised state was verified with flow cytometry to analyze the presence of

B (CD79A), T (CD3E), and NK (CD3E-/CD56+) cells.

Pigs were delivered through a sterile hysterectomy process previously described

by a board-certified large animal veterinarian [54]. Pigs were monitored regularly until

fully recovered from anesthesia and able to eat independently. Tail snips were taken

from each piglet while still under the influence of anesthesia for genotyping, and ears

were clipped for numbering. The pigs were born into sterile, gnotobiotic isolators and

were fed sterile boxed milk throughout the study to prevent infections [54].

Page 145: Determining the Oncological and Immunological Effects of

108

Human Panc01 cells (National Cancer Institute DTP, DCTP Tumor Repository)

were propagated in DMEM supplemented with 10% FBS and 1% penicillin/

streptomycin and removed from plates with Trypsin in EDTA. While the pigs were still

under the effect of anesthesia from the hysterectomy, 1.2x106 of the following human

tumor cell lines were injected: Panc01 (pancreas); U-251 (brain); HT-29 (colon); A549

(lung); or HepG2 (liver). Tumor cells were injected, subcutaneously, in 100 µL of

Matrigel behind the right ear, with an additional 100 µL of Matrigel behind the left ear.

Tumors were measured three times per week with plastic Vernier calipers. The diameter

was calculated as the square root of the product of the widest diameter and the

diameter perpendicular to that. At the same point of tumor measurement, weight and

other health monitoring was also completed. In addition to the subcutaneous cell line-

based tumors, we have also conducted orthotopic engraftment with human PDX tissues

and organoids [30, 51], significantly expanding the utility of these model animals.

4.3.5 Histotripsy Treatment of Porcine Pancreas and Liver

The ability to xenograft tumor cell lines and PDX tissue in the RAG2/IL2RG

knockout pigs orthotopically, provides a highly unique model for histotripsy

development. Our initial studies have focused on the pancreas, due in part to the high

relevance and difficulty in targeting that includes similar acoustic windows to that

expected in human patients. Initial studies have focused on refining the imaging and

targeting healthy organs with histotripsy in preparation for planned tumor ablation

studies in the RAG2/IL2RG knockout orthotopic pig model. Animals were anesthetized

Page 146: Determining the Oncological and Immunological Effects of

109

with Telazol/Ketamine/Xylazine (TKX) and maintained under anesthesia for the duration

of the imaging and histotripsy treatment. Pigs were cleaned, dried, and had the area of

interest shaved and Nair (Naircare, Ewing, NJ, USA) was applied for 10 minutes before

being washed with water to thoroughly remove hair that could interfere with imaging.

Before treatment, freehand ultrasound imaging was used to identify the desired

treatment location using both a linear ultrasound imaging probe with a frequency range

of 10-18 MHz (L18-10L30H-4, Telemed, Lithuania, EU) and a 3 MHz curvilinear imaging

probe (Model C5-2, Analogic Corp., Peabody, MA). For treatment, the histotripsy

transducer and coaxial imaging probe were mounted onto a mechanical arm with an

associated micropositioning system to provide a uniformed volumetric ablation guided

by treatment planning software (Histosonics Inc., Ann Arbor, MI) [1]. The focus of the

transducer was aligned on the imaging screen before treatment by generating a bubble

cloud in degassed water. For treatment, the transducer was grossly positioned over the

pig’s abdomen and the focus was aligned to the targeted location using ultrasound

imaging. The transducer was coupled to the pig using a degassed water bath and an

adhesive drape, as shown in Figure 2C. Once this location was identified, the

transducer was turned on and histotripsy was applied to the target using single cycle

pulses and a PRF of 500Hz, with the pressure slowly increased until a bubble cloud was

generated in the tissue. The transducer was scanned to cover a 1 cm diameter

spherical ablation volume with 500 pulses applied per treatment location. Real-time

ultrasound imaging was used to monitor the treatment throughout the procedure. After

treatment, freehand ultrasound imaging was again conducted to assess for any tissue

damage, and the animals were then immediately euthanized per protocol by an

Page 147: Determining the Oncological and Immunological Effects of

110

intracardiac injection of pentobarbital sodium and phenytoin sodium (Euthasol). A full

necropsy was conducted post-treatment by a board-certified veterinary pathologist

(S.C.O.).

4.3.6 Spontaneous Tumors in Canine Patients

In addition to the development of the SCID-like pig tumor model described above,

our team has recently initiated three studies testing histotripsy tumor ablation in

spontaneous tumors in client-owned dogs at the Animal Cancer & Research Center

(ACCRC), Virginia-Maryland College of Veterinary Medicine (VMCVM). These

prospective clinical trials evaluate the safety and feasibility of treating canine soft tissue

sarcoma and osteosarcoma tumors, which are currently underway, as well as a study

for treating canine brain tumors and others in future studies. These initial trials parallel

previous veterinary clinical trials at our site treating canine soft tissue sarcoma, liver,

brain, pancreatic, and lung cancers with either irreversible electroporation (IRE) or High

Intensity Focused Ultrasound (HIFU) thermal ablation. All of these canine tumor

populations represent potential candidates for future histotripsy studies. All clinical trials

are approved by the Institutional Animal Care and Use Committee and the Veterinary

Hospital Board. Client-owned dogs with naturally occurring tumors are recruited for

these clinical trials through the ACCRC referral network and by registry of the trial on a

publicly accessible, national veterinary clinical trials database [55]. All dogs undergo a

screening process to determine eligibility for trial enrollment, including a physical

examination, mass cytology or histopathology, complete blood count (CBC), serum

Page 148: Determining the Oncological and Immunological Effects of

111

biochemistry profile, and thoracic and abdominal imaging. Standard-of-care treatment

options are always presented to owners and dogs are only included in studies if they

meet the inclusion/exclusion criteria and if a written owner’s informed consent is

obtained. Before a study treatment is administered for our current histotripsy clinical

trials, enrolled dogs will typically undergo cross-sectional imaging of the tumor either

with contrast-enhanced CT or MRI. These imaging studies enable visualization of the

tumor characteristics as well as the surrounding and overlying tissues to assess the

available acoustic window for each study. The planned histotripsy treatment is patient-

specific and scheduled as appropriate, with all further diagnostic evaluation and

treatments performed by the Histotripsy Clinical Team at the ACCRC.

4.3.7 Histotripsy Ablation of Spontaneous Canine Tumors

The feasibility of histotripsy tumor ablation in canine patients with spontaneously-

occurring tumors was tested in two dogs, one with a peripheral soft tissue sarcoma and

one with a hindlimb osteosarcoma tumor, using the 500 kHz clinic prototype histotripsy

system (Fig.2). Patient-specific histotripsy treatment plans were designed prior to each

treatment based on pre-treatment CT imaging, physical examinations, and ultrasound

imaging assessments. These measures were used to determine the acoustic window

and the prescribed area within the tumor that was targeted with histotripsy. For

treatment, the patients were placed under general anesthesia following standard

anesthetic protocols for client-owned patients. Anesthesia was maintained with inhaled

isoflurane. The fur was shaved as closely to the skin as possible over the planned

Page 149: Determining the Oncological and Immunological Effects of

112

treatment site. Histotripsy was applied to 3cm diameter spherical volumes within the

tumor by sweeping the focal point across the target volume using the robotic micro-

positioner directed by the planning software. Vital signs (heart rate, blood pressure,

temperature, ECG, and SpO2) were monitored throughout the procedure to assess

patient safety and to identify any potential physiologic disturbances. To ensure acoustic

propagation from the transducer to the skin, a water bolus was coupled to the canine

patient over the optimal acoustic window identified using pre-treatment CT images and

freehand ultrasound imaging prior to treatment. The tumor was treated with histotripsy

under ultrasound guidance using single cycle pulses applied at a PRF of 500 Hz. Before

starting the volumetric treatment, the pressure at the focus was increased until a bubble

cloud was generated at the focus. A volumetric ablation volume was then generated

using the automated treatment strategy guided by the micropositioner and software of

the clinical histotripsy system. During histotripsy, the bubble cloud and tissue effects

were monitored using real-time US imaging. Upon completion of the treatment, patients

were recovered from anesthesia and were discharged into the care of their owners. The

patients received a physical examination and evaluation of the skin overlying the

histotripsy treatment site one day after treatment. Grading of adverse events was

determined according to the Veterinary Cooperative Oncology Group-Common

Terminology Criteria for Adverse Events (VCOG-CTCAE). A post-treatment CT scan

with contrast was performed one day after treatment to assess the completeness of the

ablation zone, the contrast enhancement pattern in the ablation zone, and the integrity

of any critical structures within or adjacent to the ablation zone. Surgical resection was

performed one day (osteosarcoma) or four days (soft tissue sarcoma) after histotripsy

Page 150: Determining the Oncological and Immunological Effects of

113

treatment. A second post-treatment CT scan was performed immediately before

resection for the soft tissue sarcoma group. The patients were placed under general

anesthesia following standard anesthetic protocols for client-owned animals to facilitate

standard surgical resection of their tumors. The histotripsy-treated area of tumor and

bone (for osteosarcoma patients), and all relevant adjacent and overlying tissue were

inspected for gross signs of damage and harvested for histological evaluation. All study

procedures complied with the NIH Guide for the Care and Use of Laboratory Animals.

4.3.8 Histology & Morphological Analysis

After treatment, tissue specimens were visually inspected and fixed in 10% formalin

for at least 24 hours before sectioning and staining. All tissues were stained using

hematoxylin and eosin (H&E) to assess for histotripsy damage and any other changes

in structure and density of collagen and other tissue structures (i.e. vessels and ducts)

within the ablation volume. To determine the estimated extent of tissue ablated within

the targeted volume, individuals trained by a board-certified veterinary pathologist

compared regions of cellular damage with regions that were outside of the ablation

zone. Results were then verified by a veterinary pathologist (S.C.O.) to ensure accuracy

and avoid biases.

Page 151: Determining the Oncological and Immunological Effects of

114

4.4 Results

4.4.1 Histotripsy Systems for Small and Large Animal Studies

Two separate histotripsy systems were used to evaluate histotripsy tumor ablation in

small (mice) and large (pigs, dogs) animal models. These separate systems were

needed to allow histotripsy to be precisely targeted to the tissues in these models based

on the tumor/tissue depths and acoustic windows available. In general, these in vivo

systems are representative of size, geometry, frequencies, and other properties of

transducers that have previously been used for applying histotripsy in small and large

animal models, respectively [2, 3, 12, 15, 20, 56].

For the small animal tumor ablation studies in the murine models, a miniature 1MHz

histotripsy transducer with a high frequency ultrasound imaging probe (Fig.1B) was

constructed to allow a well-confined bubble cloud to be generated within the shallow

subcutaneous or orthotopic tumors. This transducer was attached to a custom

motorized positioning system (Fig.1A) designed to apply automated volumetric ablations

closely mimicking those applied by the clinical histotripsy system. This transducer has a

full width half-maximum (FWHM) dimensions at a geometric focus of 0.98 mm, 0.93

mm, and 3.9 mm in transverse, elevational, and axial directions, respectively. Using this

system, a small, well-defined histotripsy bubble cloud can be generated at the focus

with approximate dimensions of ~2-3 mm in the axial directions and ~1-1.5 mm in the

transverse and elevational directions, depending on the pressure level applied. An

Page 152: Determining the Oncological and Immunological Effects of

115

example high-speed image of the bubble cloud showing the approximate dimensions of

the cloud is shown in Figure 1E. Based on the working distance of the transducer, this

system is capable of treating shallow tumors in small animal models at depths up to ~1-

1.5 cm. The smaller bubble cloud generated by this small animal system, in comparison

to clinical histotripsy devices, also allows for more precise treatments and more fine

control over ablation margins, which is necessary in order to safely and effectively

generate histotripsy ablation of murine tumors without causing off target injury to

adjacent or overlying tissues. The treatments using this system are guided and

monitored by real-time ultrasound imaging using a high frequency linear ultrasound

imaging probe aligning co-axially inside of the therapy transducer. This higher frequency

imaging (10-18 MHz) provides superior imaging quality compared to the clinical imaging

systems commonly used in histotripsy procedures, including the large animal system in

this study, allowing for high resolution images of the superficial tumor tissue, the

histotripsy bubble cloud, and the resulting tissue damage generated by histotripsy.

For the large animal studies in both pigs and dogs, a custom 500 kHz transducer

was integrated onto a clinical histotripsy system provided to our team by HistoSonics

(Fig.2). This cart-based therapy system consists of a diagnostic ultrasound imaging

system embedded onto a cart with a mechanical articulating arm and micro-positioning

system that allows for precise volumetric ablations to be generated with histotripsy

(Fig.2A). A custom 500 kHz transducer was designed with a central hole to allow for the

system’s 3 MHz curvilinear imaging probe to be coaxially aligned inside of the therapy

transducer for real-time image guidance (Fig.2B). The therapy transducer was then

mounted onto the robotic micro-positioner in order to allow histotripsy to be uniformly

Page 153: Determining the Oncological and Immunological Effects of

116

applied to a predetermined volume within the tissue, as outlined in a previous study

[15]. The transducer had FWHM dimensions at the geometric focus of 2.1 mm, 2.1 mm,

and 6.6 mm in transverse, elevational, and axial directions, respectively. Using this

system, a well-defined histotripsy bubble cloud could be generated at the focus with

approximate dimensions of ~5-6 mm in the axial directions and ~1.5-2.5 mm in the

transverse and elevational directions, depending on the pressure level applied during

treatment. An example high-speed image of a bubble cloud generated by the 500 kHz

transducer is shown in Figure 2E. Based on the working distance of the transducer, this

system is capable of treating tissue depths up to ~7 cm, allowing for treatments of the

pancreas and liver in pig models and a wide range of tumor types in canines including

superficial musculoskeletal tumors as well as abdominal cancers. Similar to the small

animal system described above, the histotripsy treatments applied using this large

animal device were all guided and monitored by real-time ultrasound imaging. However,

comparing the two systems, the large animal device provides lower resolution imaging

but more clinically relevant assessments of image-guidance for histotripsy devices that

are being developed for human applications by utilizing lower frequency imaging probes

to image at clinically relevant treatment depths in the pig and dog models that more

closely match human anatomy. Finally, it is worth noting that the larger bubble cloud

generated by the large animal histotripsy device allows for more rapid ablation of larger

treatment volumes using histotripsy while maintaining sufficient treatment precision for

targeting a predetermined volume within the target tissue.

Page 154: Determining the Oncological and Immunological Effects of

117

4.4.2 Histotripsy Treatment of Subcutaneous Murine Tumors

Although a wide variety of subcutaneous and orthotopic tumor models have been

established in mice, the majority of histotripsy studies, including the example histotripsy

treatments shown in this study have focused on subcutaneous tumor injection models

that allow straightforward assessments of tumor progression and treatment with minimal

concerns regarding acoustic windows. In general, nodular tumors developed at the

injection site for all cell line and PDX based tumors. When studying cell line based

tumors in mice, it is well characterized that most tumors tend to be less physiologically

similar to the respective spontaneous tumors in human patients. For example, the

pancreatic Pan02 tumors are densely cellular with only minimal fibrovascular stroma

(Fig.3A). The tumors often have variably-sized foci of necrosis, which typically develop

centrally within the tumor. In terms of morphology, the tumor cells typically are arranged

in vague, indistinct streams. The cells often exhibit significant pleomorphism, including

differences in nuclear size and shape across the population with numerous mitotic

figures observed throughout cells within the tumor. This is significantly different

compared to a representative primary pancreatic tumor from a human patient (Fig.3B).

We have found human patient derived xenograft (PDX) tumors in NOD scid gamma

(NSG) mice to be highly useful in reproducing the complexity and stroma of the human

primary tumor, including for studies of pancreatic cancer [45]. The PDX models have

also proved useful for studies in multiple other cancers, including the prostate, liver,

breast, and colon, shown here as a model for cholangiocarcinoma (CC) (Fig. 3C). The

representative CC image shown in Figure 3C reflects the complex, multi-cellular tumor

Page 155: Determining the Oncological and Immunological Effects of

118

with stroma and cells arranged into disorganized structures similar to de novo human

tumors.

Figure 4-3 Histotripsy Tumor Ablation in Murine Models. (A) Subcutaneous murine pancreatic Pan02 tumor. (B) Representative primary human pancreatic tumor. (C) Human CC PDX murine xenografted tumor. (D) Ultrasound images of histotripsy bubble clouds forming in RCC, (E) 4T1, and (F) CC PDX tumors. (G-I) Histology demonstrating full ablation within histotripsy targeted (G) RCC, (H) 4T1, and (I) CC PDX tumors.

Results from the histotripsy treatments of the subcutaneous RCC, PDX CC, and

orthotopic 4T1 mammary tumors showed that histotripsy was able to generate well-

defined bubble clouds within each tumor using our small animal system that were

clearly visualized on real-time ultrasound imaging (Fig. 3D-F). Using the automated

Page 156: Determining the Oncological and Immunological Effects of

119

ablation strategy, volumetric ablations of the targeted regions of the tumor were

achieved for all three tumor types (Fig. 3G-I). While the central region of the tumor

targeted with histotripsy was fully ablated, the external margins of the tumor was still

intact after treatment, similar to previous reports [19, 20, 56]. More complete ablations

of the entire tumor volume could not be achieved in the small animal models due to the

inability to treat sufficient margins around the tumor, particularly on the portion of the

tumor located immediately below the skin. When treating near these regions, prefocal

cavitation was often noted during treatment at the skin-water interface, limiting the

ability to generate a bubble cloud immediately below the skin and causing potential

cavitation damage to the skin surface, which was observed in some subjects. With the

stiffer CC tumors, the bubble cloud appeared smaller and less dynamic on ultrasound

imaging compared to the other tumor types. For this reason, the CC treatments were

applied at a higher treatment dose with finer spacing between adjacent treatment points

in order to ensure complete ablation of the targeted tumor volume. It was also noted

that there was more variability in the appearance of the bubble cloud throughout

treatment in heterogeneous tumors, with regions of suppressed cavitation. These areas

of suppressed cavitation likely correspond to the more fibrous regions of the tumor.

Overall, results showed that histotripsy could be successfully applied to all murine tumor

types tested in this study, resulting in complete ablation of the targeted central region of

the tumor with the primary limitation being the inability to treat the entire tumor volume

and a clinically relevant treatment volume around the tumor. Regardless of the

limitations, we have found that the mouse models utilized thus far for histotripsy

development, including those shown in this study, are ideal for rapid and reproducible

Page 157: Determining the Oncological and Immunological Effects of

120

parameter evaluation, hypothesis testing, and effective assessments of biological

hallmarks of cancer impacted by treatment. These models can also be used to assess

key engineering parameters necessary for device scale-up in certain cases, such as

comparing the relative differences in pulsing parameters needed to ablate different

tumor types.

4.4.3 Characterization of RAG2/IL2RG Pig Tumor Model

Our research team has previously generated and utilized immunodeficient pigs in a

variety of infectious disease and human xenograft tissue studies [57-59]. However, the

use of these pigs as cancer research models is still unique. As previously reported, we

utilized the CRISPR/Cas9 system to target the porcine RAG2 and IL2RG genes [57].

For the work described here, we utilized 454 in vitro fertilized oocytes, which were

injected with the targeted RNA for CRISPR/Cas9 gene disruption, followed by in vitro

culture [57]. Four days post-IVF, 155 embryos between the 8-cell and morula stage

were transferred to surrogate gilts. This resulted in 6 piglets being delivered by

hysterectomy into our germ-free house system, 114 days post-IVF. Genomic DNA was

collected from tail-snips for genotyping, which revealed that all 6 piglets were

successfully targeted for both the RAG2 and IL2RG genes, similar to our previous

reports [57]. Further functional verification from peripheral blood specimens, evaluated

via flow cytometry, confirmed that the piglets presented with the B-T-NK-SCID

phenotype.

Page 158: Determining the Oncological and Immunological Effects of

121

Figure 4-4 RAG2/IL2RG Immunocompromised Pig Model for Cancer Research. (A) Example of a Pan01 (pancreatic cancer) subcutaneous tumor nodule growing behind the ear and (B) once excised. (C) Control Matrigel plugs exhibits no growth. (D, G) U-251 Human Glioblastoma, (E, H) 4T1 murine breast cancer, and (F, I) HepG2 human HCC are shown by histology under low and high magnification, respectively.

For tumor engraftment, we have evaluated tumor progression of the following human

cancer cell lines in the RAG2/IL2RG knockout pigs: PANC-1 (pancreatic cancer) (Fig.

4A-B); HepG2 (liver cancer); and U-251 (brain cancer). The murine 4T1 (breast cancer)

was also engrafted. The 6 piglets were randomized and were subcutaneously injected

with 1.2x106 cells in 100 µL of Matrigel in the ear. Control injections were also added,

which were 100 µL of Matrigel alone (Fig. 4C). Each cell line was injected in triplicate in

Page 159: Determining the Oncological and Immunological Effects of

122

three separate pigs. In these initial studies, we established engraftment proficiency and

growth rates for each cell line. Within 24 hours, of the initial injection, we observed

tumor growth for all of the cell lines, except for 1, U-251 injection. This single U-251

injection was the only cell line that failed to engraft. The remaining cell lines rapidly

expanded over a 5-day period, eventually stabilizing and steadily increasing to the

target diameter of 1.0 – 1.6 cms by day 36. No changes in any clinical parameters were

noted for any of the animals. All tumors were readily palpated and observed by day 36,

whereas the Matrigel control injections were not detected. Tumors were evaluated by

ultrasound, revealing clearly defined tumors and clear delineations from surrounding

tissues. Extra-dermal, sub-dermal, and ultrasound tumor measurements were collected

at necropsy and compared for each tumor. Metastasis was also evaluated in the lung,

liver, spleen, and draining lymph nodes, with none detected. Histopathology

assessments were consistent with each tumor cell line, here showing U-251 (Fig. 4D

and G), 4T1 (Fig. 4E and H), and HepG2 (Fig. 4F and I) tumors as representative

specimens. Each was indistinguishable from the respective cell line based tumors

grown in immunocompromised mice. However, the size of the mice and the number of

tumors that can be generated is a significant limitation. This is especially true when

transitioning these studies to orthotopic tumor engraftment, where the mouse has

significant limitations in terms of human relevance. Future and on-going studies will

utilize these data to refine and optimize orthotopic tumor engraftment with these cell

lines in the RAG2/IL2RG deficient pigs. In addition to cell line tumors, we have also

successfully generated PDX-like tissue and tissue organoids from humans and

engrafted these more complex and physiologically relevant specimens into the

Page 160: Determining the Oncological and Immunological Effects of

123

RAG2/IL2RG knockout animals, opening the opportunity to move beyond cell line-based

tumor models.

Figure 4-5 Histotripsy Treatment of Porcine Pancreas and Liver. (A) Image of the water bowl and site preparation with fluid repellent adhesive surgical drape for liquid containment during histotripsy. (B) Acoustic window for pancreas ultrasound treatment identified on pre-treatment imaging. (C) Gas in the stomach and GI tract encountered prior to histotripsy treatment. (D-E) Guided by real-time ultrasound imaging, histotripsy generated a clearly defined bubble cloud in (D) liver and (E) pancreas. (F) Gross image of 0.5 cm lesion in pancreas following histotripsy. (G) Off-target effects around the stomach and GI tract. (H-I) Representative histopathology images confirm regions of ablation in the pancreas.

Page 161: Determining the Oncological and Immunological Effects of

124

4.4.4 Histotripsy Treatment of Porcine Pancreas and Liver

The feasibility of targeting the liver and pancreas in porcine subjects was tested

using the large animal histotripsy device shown in Figure 2. For this feasibility study,

pigs were imaged and treated with histotripsy in order to establish the regions of the

liver and pancreas that could be targeted for future orthotopic tumor studies in the pig

model described above. Treatments were applied non-invasively through an abdominal

window and partial ribcage obstruction (Fig.5A). The liver and pancreas were imaged

and targeted in each pig at treatment depths of ~4-6 cm (Fig. 5B-E). In the pancreas,

gas in the stomach and gastrointestinal (GI) tract was a significant issue (Fig. 5C). For

liver histotripsy treatments, well-defined bubble clouds were generated at the focus of

the transducer and clearly visualized as dynamically changing hyperechoic regions at

the focus of the transducer, as monitored by real-time ultrasound imaging (Fig.5D).

These results were consistent with previous in vivo hepatic histotripsy treatments that

have been reported in pig models [3, 11, 12, 15]. In contrast to the results seen in liver,

targeting the pancreas with histotripsy posed a more difficult challenge using our current

large animal system. Prior to treatment, the pancreas could be identified and visualized

on ultrasound imaging when using a freehand ultrasound imaging probe with significant

mechanical force applied to the abdomen to displace bowel gas (Fig. 5B-C). However,

the quality of the image was reduced significantly when the pancreas was imaged by

the co-axially aligned imaging probe within the therapy transducer (Fig.5E), likely due to

blockage from bowl gas (mechanical force couldn’t be applied in this case due to the

water standoff). In all subjects, the offset placement of the histotripsy transducer and

Page 162: Determining the Oncological and Immunological Effects of

125

imaging probe through the degassed water coupling bowl prevented the pancreas from

being clearly visualized during treatment. Based on this limitation, the focus of the

histotripsy transducer was aligned to the region expected to contain the pancreas using

nearby anatomical landmarks, and histotripsy was then applied to this as described in

the Methods. During treatment, histotripsy cavitation could be heard, but the bubble

cloud was only sporadically visualized in the focal region during treatment (Fig. 5E). In

addition, prefocal cavitation was sporadically observed during treatment in regions

outside of the focus, likely corresponding to regions containing gas-filled overlying

tissues. After treatment, gross morphology showed a small histotripsy lesion in the

pancreas, as indicated by a region of localized hemorrhage approximately 0.5 cm in

size (Fig.5F). In addition to the damage to the pancreas, off target injury to surrounding

organs, including the small intestine and stomach, was observed (Fig.5G), likely

corresponding to the regions of prefocal cavitation observed during treatment. Ablation

in the pancreas was confirmed by histopathology as a small region of localized tissue

damage inside of the pancreas (Fig.5H-I).

4.4.5 Veterinary Clinical Oncology Patient Populations for Histotripsy Development

Moving beyond small and large animal cancer models, we have also utilized

histotripsy in the veterinary clinic with translational value to human clinical studies. The

Virginia-Maryland College of Veterinary Medicine (VMCVM) Animal Cancer Care &

Research Center (ACCRC) sees a large number of canine and feline oncology patients

with a wide range of pathologies. Patients are referred from general practice

Page 163: Determining the Oncological and Immunological Effects of

126

veterinarians, veterinary specialists at external specialty hospitals, and internally from

clinical services within the VMCVM. Clinical trial participants are recruited from all

patients presenting to the ACCRC, supplemented through the Veterinary Clinical Trials

Network and Clinical Research Office (VCRO) in collaboration with the NCI

Comparative Oncology Trials Consortium (COTC)[37] and the NCI Comparative Brain

Tumor Consortium (CBTC)[60] to facilitate recruitment of patients for clinical trials. The

VCRO has a history of successfully recruiting companion animals for clinical trials and

has provided support for a number of canine and feline trials [61]. The following sections

describe the prevalence and characteristics of the most common malignancies seen at

our site, which is expected to be representative of other large veterinary hospitals in the

U.S.

In the veterinary clinic, soft tissue tumors are currently the most accessible tumor

types to treat using histotripsy and the data from subcutaneous tumors in animal models

can be directly applied. Tumors of the skin and subcutaneous tissues are common in

dogs, with malignant tumors comprising 20-40% of skin masses submitted for

histopathology [62]. Soft tissue sarcomas (STS) arise from the connective tissues,

comprising 15% of all skin tumors in dogs, and are subclassified pathologically based

on the tissue of origin [63]. Common tumor types seen in clinical practice include

fibrosarcoma, perivascular wall tumor, peripheral nerve sheath tumor, liposarcoma, and

myxosarcoma. Histologic distinction can be challenging and may be of minor clinical

importance as the majority of STS share a similar biological behavior and follow a

similar course of treatment: the patient could be cured with wide surgical excision, rarely

requiring adjuvant chemotherapy. The anatomical distribution varies, with most STS

Page 164: Determining the Oncological and Immunological Effects of

127

occurring on the trunk and limbs. Generally, STS are slow-growing and not life-

threatening. However, in veterinary practice, this often results in medical attention being

sought late in the course of disease when the curative surgical treatment option may be

limb amputation, a disfiguring surgery, or a curative surgery is no longer possible. The

range of tumor sizes seen in typical veterinary specialty practice can range from a few

centimeters up to 20 cm. Our research team has had a significant degree of success

treating STS tumors using a range of tumor ablation modalities, including HIFU which

was applied in a recent study using the Theraclion Echopulse system which is available

at our site (Fig. 6A-D).

In addition to STS, many other tumor subtypes are of interest for histotripsy,

including bone tumors. Osteosarcoma (OS) is a primary bone cancer and a devastating

disease for both human and canine patients. It is the most common primary bone tumor

in children and adolescents, as well as in the dog [64, 65]. Figure 6F shows a CT image

of an osteosarcoma in the forelimb of a dog. Treatment of OS involves removal of the

primary tumor, and chemotherapy to inhibit metastatic disease. The main cause of

death in canine and human patients with OS remains metastatic disease.

Advancements in OS therapy need to focus on treating both primary and metastatic

tumors. Dogs and humans with OS share striking similarities that make the dog an

exceptional comparative oncology research model. These similarities include matching

biological behavior, nearly identical histological features, shared global gene expression

signatures, and similar responses to standard treatment regimens [66-70]. For example,

most OS lesions in dogs and humans occur in the appendicular skeleton around the

metaphyseal region of long bones, and the disease tends to be associated with large

Page 165: Determining the Oncological and Immunological Effects of

128

and/or tall individuals [65, 66]. The anatomic similarities between the canine and human

skeletal structures allow the dog to be an accurate model for histotripsy systems

development and treatment evaluation studies. Osteosarcoma appears radiographically

identical in humans and dogs, displaying a lesion on CT and MRI scans with a

combination of lytic and proliferative bone [66, 71]. These radiographic similarities allow

for cross-species comparisons when evaluating histotripsy treatment outcomes using

imaging modalities.

The strength of naturally occurring canine OS as a model for comparative

oncology histotripsy research lies not only in the similarities of the disease in both

species but also in non-disease-related factors. The dog is an outbred species with an

intact immune system that develops spontaneously occurring OS over an extended

period, similar to humans and unlike inbred laboratory rodents. Dogs with OS reflect the

heterogeneity of human patients with OS, displaying interindividual variations in tumor

characteristics. Both humans and dogs with OS share similar clinical disease

manifestations and outcomes. The >10-fold increased incidence of canine OS

(estimated incidence of 13.9/100,000) compared to human OS (1.02/100,000), the

shorter lifespan of dogs, the rapid progression of metastatic OS in dogs enable

outcomes to be assessed in relatively short periods compared to their human

counterparts [66, 72, 73]. Metastatic bone tumors also occur in dogs, and common

primary cancers that metastasize to bone include mammary carcinoma, prostatic

carcinoma, urogenital carcinoma. Osteosarcoma can also metastasize to bone. The

ACCRC treats approximately 25 canine patients with OS annually, and this number is

expected to increase with recruitment for clinical trials. Histotripsy, with its ability to

Page 166: Determining the Oncological and Immunological Effects of

129

disintegrate solid tumors and its potential for immunogenic stimulation, is uniquely

poised to have the potential to achieve the goals of OS therapy – treating both the

primary tumor and metastatic lesions.

Liver cancer is a preferred target for histotripsy and is the focus of the first human

clinical trial of the ablation strategy in patients [18]. Primary liver cancer in humans is

the fifth most common cause of cancer death in men and the ninth most common cause

of cancer death in women. Over the past two decades, incidence rates have increased,

and the overall mortality rates have been rising an average of 2.6 percent each year

[74]. At present, only 10-23% of human patients with HCC are surgical candidates for

curative-intent treatment [75, 76]. Liver transplantation is the only therapeutic option that

offers a consistent 90% survival rate, but the scarcity of liver donors severely limits its

broad applicability [77]. Primary liver tumors in dogs are considered infrequent and

account for 1.5% of all canine tumors, with hepatocellular carcinoma (HCC) being the

most common (Fig. 6G) [78]. The prognosis is good for massive HCC when surgical

resection is possible [79]. In contrast, the prognosis is poor for dogs with malignant

tumors other than massive HCC, and especially for dogs with nodular and diffuse HCC,

as surgical resection is not possible. Literature suggests that 40% of canine HCC are of

nodular or diffuse presentation and that a significant proportion of the massive HCC

tumors are non-resectable with complete margins due to their hilar or bile duct

association. Bland embolization and chemoembolization have been reported with

variable success in the palliation of four dogs with HCC [80].

Page 167: Determining the Oncological and Immunological Effects of

130

Figure 4-6 Example Spontaneous Tumors in Canines. (A) Soft Tissue Sarcoma (STS) presented as a firm, fixed, painful soft tissue mass at the right proximal forelimb of a dog. (B) Contrast-enhanced CT consistent with a soft tissue attenuating and moderately contrast enhancing mass (yellow asterisk). The mass lies lateral to the triceps muscle and arises from the ascending pectoral muscle. (C) HIFU treatment of the tumor to partial ablation. An Echopulse unit by Theraclion was used. (D) Gross appearance of a cross-section of the tumor, 4 days post HIFU treatment. The yellow arrows point to the ablated tumor volume. (E) CT image of a rostral maxillary tumor in a dog, indicated in circled area. Note the aggressive invasion of the tumor into the maxilla, causing bone lysis. (F) CT image of a humeral osteosarcoma in a dog, indicated by the arrow. Note the aggressive nature of the tumor, causing bone lysis and proliferation. (G) CT image of a liver tumor in a dog, indicated by the circle. (H) CT image of a pancreatic tumor in a dog, indicated by the arrow. (I) CT image of a metastatic lymph node in the same patient, indicated by a circle.

Page 168: Determining the Oncological and Immunological Effects of

131

In situ, energy-mediated tumor ablation is an alternative therapy to resection and

can be performed using a minimally invasive approach (percutaneous or laparoscopic)

[81]. Thermal ablation (radiofrequency and microwave ablation [RFA/MWA]) are the

mainstays of liver tumor ablation [81], with MWA currently superseding RFA as the

technology of choice [82]. Thermal ablation has similar oncological outcomes to

resection in carefully selected patients [83, 84]. However, it results in indiscriminate

tissue destruction at the near-field ablation zone, inducing a significant risk for

excessive, non-tumor tissue loss and damage to biliary and vascular structures,

resulting in high rates of liver-related complications [82, 85, 86]. Thermal HIFU has

enabled ablation of liver nodules and total tumor ablation is multiple studies for selected

human patients, but can also result in off-target effects to biliary and vascular structures

[87-89]. Thus, non-thermal ablative approaches are gaining interest in the treatment of

primary and metastatic liver tumors. Non-thermal ablative methods may also present a

unique opportunity to harness the patient’s immune system, as part of a multimodality

therapeutic approach. Rodent data suggest that tumor ablation by non-thermal

mechanisms can lead to anti-tumor immunity [90-92]. The liver represents an

immunologically rich and active organ, with intrahepatic immune cells skewed toward

tolerance, with quiescent or overtly suppressive functions [93, 94]. By attacking cancer

using non-thermal ablation, the tumor has the potential to serve as its own multivalent

anti-cancer vaccine through the presentation of tumor antigens and damage associated

signals originating from the ablated tissue [95]. At ACCRC, we have performed

approximately 15 HCC resections, about 8 liver tumor ablations annually, and we

Page 169: Determining the Oncological and Immunological Effects of

132

evaluate an equal number of non-resectable cases. We demonstrated the feasibility and

safety of non-thermal liver tumor ablation using High-Frequency Irreversible

Electroporation (H-FIRE) by completing the first-in-patient pilot study of percutaneous

H-FIRE in dogs diagnosed with HCC. CT imaging before and 4 days after H-FIRE

indicated a predictable tumor ablation volume, and analysis of the treated tumor tissue

indicated specific T-cell recruitment and gene expression signatures associated with cell

injury/death and cell-mediated immunity [96]. Histotripsy represents an attractive, non-

invasive, non-thermal approach for the ablation of liver nodules. Histotripsy’s precise

ablation targeting capabilities in combination with the completely non-invasive approach

and the ACCRC’s experience in minimally invasive ablative approaches for liver cancer

would allow for the canine cancer patients diagnosed with liver cancer to serve as the

translational model for its application as an outpatient procedure.

Similar to humans, pancreatic cancer in dogs is also a critically important

malignancy. Pancreatic tumors in dogs can originate from the exocrine pancreas, for

example, adenocarcinomas of the pancreatic ducts or acini, or originate from the

endocrine pancreas, for example, insulinoma. Insulinoma is the most common

pancreatic endocrine tumor in dogs, originates from pancreatic cells and secretes

insulin, causing potentially life-threatening hypoglycemia [97]. Insulinoma also

aggressively metastasizes to other organs, commonly to the liver and regional lymph

nodes. Figure 6H-I shows CT images of a pancreatic tumor with its associated lymph

node metastasis in a dog. Canine insulinomas behave in a biologically similar fashion to

human malignant insulinoma [98]. The functional insulin-secreting nature of primary and

Page 170: Determining the Oncological and Immunological Effects of

133

metastatic insulinoma is a major life-limiting factor for patients, and treatment of both

primary and metastatic disease is essential for improving prognosis.

Surgical resection of all gross disease in dogs with pancreatic cancer, though not

curative, is recommended to improve the efficacy of medical adjuvant treatments and

disease outcomes [97]. Complete surgical resection of all gross disease is not always

achievable, depending on the location and extent of the lesions. Resection of tumors

located in the body of the pancreas is associated with high morbidity and potentially

mortality rates, and the extent of metastatic lesions may preclude surgical resection.

Novel treatment approaches are needed to improve the long-term survival of pancreatic

cancer patients. New treatment techniques that can non-surgically ablate tumor lesions

are attractive alternates to surgical tumor resection. Treatment approaches to stimulate

the host immune system to mount an anti-tumor immune response that carries immune

memory will improve the prognosis for the development or recurrence of metastatic

disease in pancreatic cancers. Histotripsy, with its ability to precisely ablate solid tumors

and its potential for immunogenic stimulation, is uniquely poised to advance the

therapeutic efficacy and prognosis of insulinomas, a tumor with currently limited

treatment options. The ACCRC treats approximately 8 canine patients with insulinoma

annually, and this number is expected to increase with recruitment for clinical trials. The

ACCRC has treated insulinoma tumors in dogs using another investigational non-

thermal tumor ablation technique, H-FIRE, and will continue its investigations into

pancreatic tumor ablations using histotripsy.

Canine oral tumors are another tumor type that can serve as strong comparative

oncology research models for their human counterparts. For instance, canine oral

Page 171: Determining the Oncological and Immunological Effects of

134

melanoma has been shown to represent a faithful model for human mucosal melanoma

and human triple wild-type melanoma [99-101]. The disease in both species shares

genetic signatures, clinical behavior, and histological characteristics [102, 103]. Head

and neck squamous cell carcinomas are the most common oral cancer in people, and

canine oral squamous cell carcinoma shares similarities with the human form of this

disease [104]. Patients with oral cancers commonly present with clinical signs

associated with pain due to the oral cancer, decreased appetite, and weight loss. Oral

tumors in dogs occur with relatively low frequency, constituting 6% of all canine

cancers, and three of the most common oral cancers in dogs are malignant melanoma,

squamous cell carcinoma, and fibrosarcoma [105-107]. Oral osteosarcoma (OS)

represents 12 - 13% of all canine OS [108]. Canine oral tumors are commonly locally

invasive and many are aggressively metastatic, especially to the locoregional lymph

nodes. A non-surgical tumor ablation technique such as histotripsy, which is also

capable of stimulating an anti-tumor immune response and can be used for tissue

selective ablation of tumors near critical structures, has exciting potential to benefit both

canine and human patients suffering from oral cancers. The ACCRC treats

approximately 8-10 canine oral tumors annually. Figure 6E shows an example of a

canine oral tumor that is locally invasive, causing bone lysis of the rostral maxilla.

Our research team has been highly successful in utilizing tumor ablation

modalities to treat canine brain tumors [109]. Despite significant progress over the last

decade, brain tumors such as malignant gliomas (MG), represent some of the most

treatment-refractory cancers in both humans and dogs, and local treatment failures

remain a significant source of morbidity and mortality in brain tumor patients. Dogs and

Page 172: Determining the Oncological and Immunological Effects of

135

humans are the only mammals known to spontaneously and commonly develop MG,

and MG accounts for approximately 35-40% of all primary brain tumors in dogs. As

dogs have a complex gyrencephalic brain, and canine gliomas share many clinical,

imaging, histomorphologic, molecular, and genetic features with human gliomas, dogs

with brain tumors are uniquely suited translational model systems for the study of tumor

ablation strategies [90].

In both dogs and humans, there is a dire need for new approaches that can be

applied to tumors involving eloquent brain regions, circumvent or disrupt the blood brain

barrier, target highly resistant tumors and cancer stem cells, generate minimal collateral

damage to healthy tissue, and promote an anti-tumor immune response. Successful

therapeutic strategies are likely to require a combination approach involving surgery,

chemo and radiation therapy, and novel ablation approaches that can de-bulk the

primary tumor and attenuate recurrence driven from residual cancer stem cells

repopulating the resected tissue, or predictably and reversibly disrupt the blood brain

barrier in a penumbra of tissue surrounding the bulk tumor to facilitate the delivery of

therapeutics that are normally impermeant to the brain [110]. Histotripsy has been

utilized in pre-clinical porcine models to ablate brain tissue. However, it is still in the

early stages of development for the treatment of brain tumors in the veterinary clinic.

Unlike many of the other tumors discussed thus far, a significant challenge to the

application of histotripsy in the brain is the thickness inhomogeneities inherent to the

skull, which can cause aberrations in transcranially propagated ultrasound pulses

resulting in reduced focal pressure amplitudes in the target tissue. Thus, a craniectomy

is required to provide an acoustic window for effective bubble cloud formation. To

Page 173: Determining the Oncological and Immunological Effects of

136

overcome current obstacles associated with histotripsy in the brain, our research team

has developed novel acoustically transparent cranial implants and is rapidly adapting

clinical histotripsy systems for use in active clinical trials in dogs with brain tumors.

4.4.6 Histotripsy Ablation of Spontaneous Canine Tumors

Figure 4-7 Histotripsy Treatment of Canine Soft Tissue Sarcoma. (A-B) Image of spontaneous soft tissue sarcoma with estimated tumor size of ~7-8 cm in diameter. (C) Contrast-enhanced CT collected before histotripsy treatment. (D) Histotripsy treatment experimental set-up. (E) Real-time ultrasound image collected during histotripsy treatment showing the bubble cloud visible as a hyperechoic, oscillating zone (arrow). (F) Contrast-enhanced CT collected 4 days after histotripsy treatment with decreased contrast uptake visible in the zone of ablation. (G) Gross morphological analysis of resected canine soft tissue sarcoma. Box indicates treatment region, identified by location and presence of tissue necrosis and hemorrhage. (H) H&E image of untreated canine soft tissue sarcoma (20x magnification) with intact cells and undisturbed extracellular matrix is observable. (I) H&E image of canine soft tissue sarcoma tissue treated with histotripsy (40x magnification) showing extensive necrosis and hemorrhagic appearance.

Page 174: Determining the Oncological and Immunological Effects of

137

Figure 4-8 Histotripsy Treatment of Canine Osteosarcoma. (A-B) Image of an undisturbed spontaneous canine osteosarcoma in the left hind limb. Significant soft tissue swelling was present, creating an estimated zone of ~30 cm of affected tissue. (C) Contrast-enhanced CT collected before histotripsy treatment. (D) Histotripsy treatment experimental set-up. (E) Real-time ultrasound image collected during histotripsy treatment with the bubble cloud visible as a hyperechoic, oscillating zone (arrow). (F) Contrast-enhanced CT collected 1 day after histotripsy treatment.

The feasibility of conducting in vivo histotripsy treatments in canine patients with

spontaneous tumors was tested in two initial patients with a peripheral soft tissue

sarcoma (Fig.7) and a hindlimb osteosarcoma tumor (Fig.8). Histotripsy was applied

non-invasively at a p- estimated to be ~30-35 MPa. A 3 cm spherical ablation volume

was targeted inside of both tumors, and histotripsy was applied uniformly over this

volume at a PRF of 500 Hz and a dose of 500 pulses/point, with a total treatment time

of ~59 minutes. In both dogs, well-defined histotripsy bubble clouds were generated at

the focus of the transducer and clearly visualized as dynamically changing hyperechoic

regions throughout the treatment, as monitored by real-time ultrasound imaging (Fig.7E,

Page 175: Determining the Oncological and Immunological Effects of

138

Fig.8E). The procedures in both dogs were tolerated well throughout treatment, with no

signs of distress or changes in vital signs during the procedures. After treatment, both

subjects were recovered until follow-up CT imaging and surgical resection one day

(osteosarcoma) or 4 days (soft tissue sarcoma) after treatment. Both the pre- and post-

treatment CT images (Fig.8C/F) for the dog with the osteosarcoma tumor did not show

a clearly defined tumor or ablation zone after treatment, likely due to the heterogeneous

nature of the osteosarcoma tumor, the presence of significant edema, or CT imaging

parameters. Future work will aim to improve the CT imaging for these patients or utilize

MRI imaging, as needed. The post-treatment CT image for the dog with the peripheral

soft tissue sarcoma showed a clearly defined ablation zone closely matching the region

targeted with histotripsy (Fig.7F). The ablation zone can be clearly visualized on CT as

a darkened region of the tumor cross-sectional image (Fig.7F), indicating a decrease in

uptake of contrast and blood flow to this region which correlates with the successful

histotripsy treatment. After treatment, gross morphology showed a histotripsy lesion was

generated in the targeted tumors, and the tissue was processed for histological analysis

after resection. Figure 7G-I shows an example of gross morphology and histology for

the peripheral soft tissue sarcoma tumor treated with histotripsy, with results showing

histotripsy generated complete ablation of the targeted tumor tissue. Together, results

from these initial treatments provide an example of histotripsy tumor ablation in dogs

with spontaneous tumors and demonstrate the potential of utilizing veterinary oncology

populations for conducting more clinically relevant studies of histotripsy for cancer

applications.

Page 176: Determining the Oncological and Immunological Effects of

139

4.5 Discussion

While early diagnosis and multi-scale combinatorial treatment approaches are

essential to reducing cancer related mortality, the lack of treatment options for many

types of cancers has resulted in an unacceptable stagnation in patient morbidity and

survival. New treatment options and therapeutic strategies are critical to improve the

survival of these patients. Focal tumor ablation modalities have shown recent success

in multiple clinical trials by crossing barriers that are difficult to bridge by chemotherapy

and surgery. Histotripsy is emerging as one of the more promising modalities. However,

as with all of the other tumor ablation strategies, clinical translation for many of the most

difficult to treat malignancies has been hampered by the lack of robust pre-clinical

animal models. While some modalities, such as radiotherapy and microwave ablation,

have the historical advantage of multiple years of clinical data in human patients, it is

much more difficult for emerging technologies, such as histotripsy or IRE, to be gain

widespread adoption in the human clinic without more mechanistic insight that defines

their therapeutic advantages. Indeed, the non-thermal nature of these modalities

appears to have significant benefits and increasingly nuanced mechanisms that require

more robust assessments to fully understand their mechanisms of action. For example,

it is becoming increasingly apparent that these modalities are significantly better than

currently adopted strategies at engaging the immune system following treatment [19,

92]. This has significant implications associated with the so-called “abscopal effect”

[111], which appears to occur more predictably and robustly following non-thermal

ablation. This has multiple benefits, including reduced metastatic burden, lower

Page 177: Determining the Oncological and Immunological Effects of

140

recurrence rates, improved responsiveness to immunotherapeutics, and improved

overall survival. However, the improved systemic effects associated with local tumor

ablation significantly increases the complexity of the studies necessary to validate these

mechanisms. Thus, necessitating the development of improved pre-clinical animal

models.

In terms of animal models, the use of rodents in tumor ablation studies continues

to be widely accepted. This is certainly understandable due to the historic use of mice

and rats in the cancer biology and biomedical engineering fields. Indeed, we have

already mentioned several advantages and disadvantages, such as the small size that

can be highly detrimental for engineering and device design. This often necessitates the

miniaturization of devices that work well in rodents but either fail to scale up and

translate well to humans or require additional validation in large animal studies prior to

regulatory approval for human testing. For example, the histotripsy system used to treat

the mouse tumors in this study consisted of a highly customized therapy transducer that

could only target small, superficial tumors at depths <1.5 cm. Using this system to treat

tumors in mice does not answer key questions about the potential feasibility of treating

tumors in human patients through the acoustic windows available for a given pathology

or the optimal approaches for achieving complete volumetric ablation of human tumors

(with adequate clinical margins) that are located in different regions and with a wide

range of characteristics including tumor size, the number of nodules, and the presence

of critical structures within or near the tumor. Furthermore, in addition to the limitations

of the therapy transducer, the small animal system also utilizes a high frequency

ultrasound imaging system that provides excellent real-time treatment guidance and

Page 178: Determining the Oncological and Immunological Effects of

141

monitoring but only provides minimal information about the potential to guide and

monitor histotripsy treatments in future clinical studies. Although small animal tumor

studies are often supplemented with large animal experiments in healthy animals, such

as pigs, studies of histotripsy ablation of healthy tissue do not allow for investigations

into the effectiveness of histotripsy to achieve precise and complete ablation of a target

tumor volume nor the ability to assess real-time and post-treatment imaging methods to

separately visualize the ablation zone from residual untreated tumor. Finally, these

limitations make it difficult to assess the safety of histotripsy for treating larger tumors

that are typically seen in humans as well as the safety of ablating tumors in an animal

with similar anatomy and physiology to human patients.

In many ways, this failure to translate new ablation methods, such as histotripsy,

into the clinic is associated with the choice of cancer models used to evaluate, validate,

and optimize therapeutic strategies. For example, most tumor ablation studies have

taken advantage of subcutaneous tumor engraftment models, especially during early

modality development. As discussed in our results section, these models have several

advantages and are highly useful, especially during early proof-of-concept/proof-of-

principle testing. The subcutaneous engraftment of human cancer cell lines in

immunocompromised mice, for example in the flank, have a long history of use in

studying cancer hallmarks. In terms of histotripsy, these models offer a readily

accessible tumor where the direct effects of ablation can be evaluated, both visually and

by ultrasound. Likewise, the use of human cells in the mouse allows for straightforward

visualization of ablation using human and mouse-specific antibodies and other reagents

to label the human-derived tumor cells and host specific mouse cells. Unfortunately, as

Page 179: Determining the Oncological and Immunological Effects of

142

shown and discussed in our results, tumors generated subcutaneously and using cell

lines have several disadvantages. Specifically, the tumor morphology, biology, and

stroma are significantly altered from the original host tumor. In the context of histotripsy,

this is a significant concern where tissue characteristics, particularly tissue fibrosis and

other factors that impact the local tissue mechanical strength, can significantly impact

ablation success [6, 14, 112-114]. Without adequate models that can recapitulate the

relevant tumor microenvironment, it is difficult to identify the optimal histotripsy

parameters and treatment doses needed to achieve complete tumor ablation or to

develop improved histotripsy strategies for developing tumor-selective ablation methods

that can ablate the tumors while preserving critical structures such as vessels, nerves,

and bile ducts [1, 3, 9, 13, 14]. Likewise, the lack of a functional immune system does

not allow the evaluation of this increasingly important aspect of the tumor

microenvironment that may have significant implications in histotripsy effectiveness.

The use of rodent derived cancer cells in the host species is a significant

improvement over the use of human cell lines in immunocompromised mice. These cell

lines are more amenable to orthotopic studies, where the cells can be engrafted into the

tissue of relevance in immunocompetent mice. This provides significantly increased

relevance, a more accurate host tissue stroma, and the intact immune system allows for

assessments of both local and systemic anti-tumor responses following treatment,

which is becoming more essential based on recent studies showing the potential

immunological benefits of histotripsy. The ability to genetically modify both the host and

the cancer cell line also allows for robust mechanistic studies. It should be noted that

the mouse strain and sex are both critical factors to consider in these models. For

Page 180: Determining the Oncological and Immunological Effects of

143

engraftment to be successful, the strain (i.e. C567Bl/6 or BALB/c) of the host and the

cell line must match or the host immune system will reject the tumor. Likewise, male

derived tumor cells can fail to engraft in female hosts due to Histocompatibility Y antigen

incompatibility and tissue rejection. Thus, while the intact immune system is an

advantage, this can limit tumor models in rodents. Likewise, similar to the limitations

discussed above for human cell lines and as shown in our results here, the same

limitations associated with unrepresentative tumor microenvironments, lack of cell

diversity within the tumor, and inaccurate tumor-associated stroma all exist in mouse

cancer cell line derived tumors. Each of these are major issues for histotripsy

development.

To circumvent many of these limitations, our research team has recently

deployed PDX models for histotripsy development [45]. PDX rodent models utilize the

engraftment of cancer tissue, typically biopsy or surgically resected tissue fragments,

into immunocompromised mice. These mice are typically NOD scid gamma (NSG)

mice. As shown and discussed in our results section, over time these tumor fragments

will proliferate into a tumor that closely mimics the biological and stromal complexity of

the original patient’s tumor. The tumor can be expanded to provide ample tissue for ex

vivo/in vitro studies necessary for device development, the tissue can also be evaluated

in the animal. Likewise, the added ability to detect human cells in mice can provide high

resolution assessments of tumor ablation. While this method still suffers from the lack of

an immune system niche, this strategy is incredibly useful for the evaluation of tumor

specific ablation to define parameters that are impacted by unique tumor

microenvironments, cell complexity, and stroma characteristics.

Page 181: Determining the Oncological and Immunological Effects of

144

Moving beyond mice, large mammal pre-clinical models can also be effectively

incorporated into tumor modality development. Here, we propose the incorporation of

pigs due to their overall anatomical and physiological similarities to humans. Likewise,

as we discuss in the results above, genetic manipulation is now becoming more

commonplace and the pig is emerging as a key cancer model due to the development

of the immunocompromised RAG2/IL2RG deletion animals that are receptive to cancer

cell engraftment and the rise of genetically modified animals, such as the “oncopig” [29].

In terms of histotripsy development, we have focused on the RAG2/IL2RG deletion pigs.

These animals are functionally similar to the NSG mice, mentioned above, and we have

found that they can be used in identical applications. Advantages of this model include

the ability to surgically implant human (or other species) cancer cell lines in specific

locations within organs of interest, such as adjacent to major blood vessels or nerves.

The cell lines have a predictable growth rate when implanted either subcutaneously or

in situ, allowing for a highly reproducible and effective model system to evaluate tumor

ablation modalities. It should be noted that the use of the CRISPR/Cas9 system allows

for the generation of animals “on-demand”, without the need for maintaining breeding

colonies. However, one disadvantage is that the targeting of the RAG2/IL2RG genes

are less predictable. For example, as mentioned in the results section, one of the U-251

cell lines did not successfully engraft in the pigs. Further genotyping of this animal

revealed only a partial knockdown of the RAG2 and IL2RG genes, resulting in an

attenuated but still viable immune system that drove tumor rejection. Thus, before

orthotopic injection, it is critical to fully determine both genotype and immune system

phenotype in these animals. It is also important to note that due to their

Page 182: Determining the Oncological and Immunological Effects of

145

immunocompromised status, these animals require housing under germfree conditions

that are only available at a small number of institutions worldwide. Moving beyond

human cell line studies in this unique model, which have some of the same limitations

discussed above for the mouse studies, these animals are uniquely amenable to more

complex human tissue engraftments, such as from PDX tissues and organoids. Thus,

as we move into the future, these highly useful specimens will allow for orthotopic

engraftment in these unique animals for more robust histotripsy development,

refinement, and treatment optimization.

We have successfully incorporated porcine models into our tumor ablation

modality testing pipeline. In addition to the tumor models discussed above, healthy pig

models have already proven useful in defining histotripsy treatment parameters utilized

in current and planned clinical trials [15, 18]. For example, as we report here, we have

successfully refined the use of histotripsy in targeting the healthy liver in pigs. By

expanding these studies to include liver tumors in the pig models, we expect to take

advantage of the features of healthy pig models that are anatomically similar to humans

while also being able to compare tumor-specific therapy and imaging methods that can’t

be tested in healthy animals, such as the safety and effectiveness of ablating liver

tumors located near critical structures or the ability to visualize and target liver tumors in

locations that require acoustic access through partial or complete rib coverage. In

addition to the liver, the work in this study has started to extend the use of histotripsy as

a non-invasive method for ablating pancreatic tumors. In the results presented above,

we report here for the first time histotripsy being used to target the pancreas in a large

animal model. This is an important milestone, but also illustrates some of the difficulties

Page 183: Determining the Oncological and Immunological Effects of

146

in treating the pancreas and further supports the use of large animal models, such as

the pig, in device development. The acoustic window is critical to most applications of

histotripsy, including pancreatic cancer, and it cannot be effectively replicated in mouse

models. As we report here, gas in the stomach and GI tract can significantly limit the

ability to visualize the pancreas during histotripsy procedures and may also limit

histotripsy from generating a precise and effective bubble cloud in the pancreas without

causing off-target cavitation injury. We have recently developed highly effective

protocols to better alleviate these issues, which range from engineering changes to the

device to refining our imaging and treatment parameters for use in pancreatic cancer,

as well as, investigating other changes such as diet adjustments, pharmaceutical

interventions, and surgical/interventional strategies to remove excess gas prior to

treatment. For example, we have successfully utilized the addition of custard to the pigs’

diet and optimized a cocktail of simethicone (anti-gas) and Bisacodyl (laxative) as a less

invasive strategy to improve the acoustic window, similar to studies reported in human

patients preparing for HIFU treatment [115, 116]. Ultimately, our histotripsy treatment

parameters will be combined with our tumor models described above. Consistent with

ongoing human and veterinary clinical studies, we anticipate that tumors in the

pancreas will be easier to image and target compared to the healthy pancreas. Moving

beyond the pancreas, in this report, we describe the use of multiple cell lines from

cancers currently being evaluated by our research team. In the future, we plan to

incorporate histotripsy data generated here from the liver and pancreas to refine the

treatment parameters of these other cancers and tissues, defined here in subcutaneous

models, including the use of PDX specimens and organoids to provide an even more

Page 184: Determining the Oncological and Immunological Effects of

147

robust and translational model for both human and veterinary patients with a wide range

of cancer types.

In addition to human applications, tumor ablation modalities such as histotripsy

are also emerging therapeutic strategies in the veterinary clinic, especially in veterinary

oncology. Ablation techniques such as histotripsy offer an attractive alternative

treatment to surgical resection of tumors. The current standard-of-care for many tumors

in veterinary oncology includes surgical resection of the primary tumor. However,

surgical resection sometimes is accompanied by excessive risk and morbidity. For

example, limb salvage surgery in dogs to resect the primary tumor in OS is associated

with a high rate of complications, with major complication rates as high as 71% reported

[117]. Additionally, despite the advancements in chemotherapeutics, many veterinary

cancer patients, like their human counterparts, eventually succumb to metastatic

disease. Histotripsy, with its ability to disintegrate solid tumors and its potential for

immunogenic induction of an anti-tumor response, is uniquely poised to achieve the

major goals of cancer therapy – treating both the primary tumor and metastatic lesions.

Not only can veterinary cancer patients directly benefit from novel ablation technologies

such as histotripsy, they also provide excellent clinical trial populations for comparative

oncology studies to benefit humans. The anatomic, physiologic, and cancer biology

similarities between many of the animal species treated in the veterinary clinic to human

cancer patients offer a direct translation of technologies from bench-to kennel-to

bedside [118]. For example, the results from the histotripsy treatments of canine STS

and OS shown in this study highlight many of the benefits of testing histotripsy in dogs

with spontaneous tumors. More specifically, treatments were conducted with a clinical

Page 185: Determining the Oncological and Immunological Effects of

148

prototype histotripsy system that consists of the therapy transducer, imaging system,

and volumetric ablation treatment strategies that are similar to those expected to be

used in humans. The results from these studies, which demonstrated histotripsy could

achieve precise and complete ablation of targeted volumes within both the STS and OS

tumors, are expected to be more directly translatable to humans when it comes to the

devices and treatment parameters used for histotripsy. In addition to providing a more

clinically relevant population for testing the safety and efficacy of histotripsy devices, the

dog as a comparative oncology research model also offers the unique advantage of

being an outbred species with an intact immune system that develops spontaneously

occurring tumors over an extended time, just as humans do, unlike inbred laboratory

rodents that are not syngeneic host-tumor models. Utilizing canine models of

spontaneous disease allows for evaluation of immune responses to treatments, an

evaluation that is not as readily achieved in immunodeficient rodent models. Dogs and

humans are more alike anatomically and physiologically than are mice to humans and

share the same physical environments, unlike laboratory rodents maintained in a highly

controlled research facility environment [70]. Dogs with spontaneously occurring tumors

represent powerful tools for device development and clinical trials, as they are a

biologically relevant disease model that can serve to better bridge preclinical murine

study findings with human clinical trials. Additionally, the willingness of canine owners to

engage in sample collections and procedures such as biopsies make dogs an

invaluable model for clinical testing. Conversely, client-owned dogs present limitations

such as the low prevalence of certain tumor types, for example, intestinal cancer,

compared to humans, thus limiting the study of these diseases.

Page 186: Determining the Oncological and Immunological Effects of

149

4.6 Conclusion

Histotripsy is rapidly emerging as the first completely non-invasive, non-thermal, and

non-ionizing ablation method for the treatment of cancer. However, the lack of clinically

and physiologically relevant pre-clinical cancer models has slowed the translation of

histotripsy into human clinical trials. This paper highlights the pros and cons of a diverse

range of small and large animal tumor models, including spontaneous tumors in

veterinary patients that are currently being developed for investigating histotripsy for

oncology applications. Results of example histotripsy treatments performed in mouse

tumor models demonstrate the feasibility of using mice as highly cost-effective models

that can test histotripsy for a nearly limitless range of cancer types. However, results

also show the limitations of mouse tumor models due to their small size, lack of clinical

and physiological relevance to humans, and the inability to test clinically relevant

histotripsy devices and treatment protocols. To address these limitations, our team has

developed a novel RAG2/IL2RG deletion pig tumor model that is being developed for

histotripsy studies. Results from preliminary studies using this model show its potential

to be used with a wide range of tumor types to study histotripsy tumor ablation in a

clinically relevant large animal model. Preliminary histotripsy experiments in the healthy

pig liver and pancreas further demonstrated the strengths of this model, which will be

utilized in planned studies testing histotripsy pancreatic tumor ablation using orthotopic

tumors grown in the pig pancreas as well as future studies with additional cancer types.

Finally, this work highlights the potential benefits of developing histotripsy for the

treatment of spontaneous tumors in veterinary patients and utilizing these veterinary

Page 187: Determining the Oncological and Immunological Effects of

150

patient populations to provide more realistic models for developing and testing

histotripsy devices and techniques designed for future use in human patients. Together,

these studies demonstrate the potential advantages of bench-to kennel-to-bedside

approaches in the development of histotripsy for oncology applications that should be

considered to improve the rate and success of efforts to translate histotripsy into clinical

practice for the treatment of cancer.

4.7 Acknowledgment

This work was supported by grants from the Focused Ultrasound Foundation (FUSF-

RAP-S-18-00011, FUS746), The National Institutes of Health (R21OD027062,

R21EB028429, R21EB030182, R21EB027979), Grayton Friedlander Memorial Fund,

and the American Kennel Club Canine Health Foundation. This work was also

supported by the Virginia Maryland College of Veterinary Medicine; The Virginia Tech

Institute for Critical Technology and Applied Sciences Center for Engineered Health;

and the Virginia Tech Department of Biomedical Engineering and Mechanics.

Page 188: Determining the Oncological and Immunological Effects of

151

4.8 References

[1] C. M. Smolock AR, Vlaisavljevich E, Gendron-Fitzpatrick A, Green C, Ziemlewicz

TJ and Lee FT, "Robotically Assisted Sonic Therapy as a Non-Invasive Non-

Thermal Ablation Modality: Proof of Concept in a Porcine Liver Model,"

Radiology, vol. 287, no. 2, pp. 485-93, 2018.

[2] E. Vlaisavljevich et al., "Non-Invasive Ultrasound Liver Ablation Using Histotripsy:

Chronic Study in an In Vivo Rodent Model," Ultrasound Med Biol, vol. 42, no. 8,

pp. 1890-902, Aug 2016, doi: 10.1016/j.ultrasmedbio.2016.03.018.

[3] E. Vlaisavljevich et al., "Image-guided non-invasive ultrasound liver ablation

using histotripsy: feasibility study in an in vivo porcine model," Ultrasound Med

Biol, vol. 39, no. 8, pp. 1398-409, Aug 2013, doi:

10.1016/j.ultrasmedbio.2013.02.005.

[4] T. Worlikar et al., "Histotripsy for Non-Invasive Ablation of Hepatocellular

Carcinoma (HCC) Tumor in a Subcutaneous Xenograft Murine Model," Conf Proc

IEEE Eng Med Biol Soc, vol. 2018, pp. 6064-6067, Jul 2018, doi:

10.1109/EMBC.2018.8513650.

[5] A. D. Maxwell, C. A. Cain, T. L. Hall, J. B. Fowlkes, and Z. Xu, "Probability of

cavitation for single ultrasound pulses applied to tissues and tissue-mimicking

materials," Ultrasound Med Biol, vol. 39, no. 3, pp. 449-65, Mar 2013, doi:

10.1016/j.ultrasmedbio.2012.09.004.

Page 189: Determining the Oncological and Immunological Effects of

152

[6] E. Vlaisavljevich et al., "Effects of ultrasound frequency and tissue stiffness on

the histotripsy intrinsic threshold for cavitation," Ultrasound Med Biol, vol. 41, no.

6, pp. 1651-67, Jun 2015, doi: 10.1016/j.ultrasmedbio.2015.01.028.

[7] Z. Xu et al., "Controlled ultrasound tissue erosion," IEEE Trans Ultrason

Ferroelectr Freq Control, vol. 51, no. 6, pp. 726-36, Jun 2004, doi:

10.1109/tuffc.2004.1308731.

[8] R. M. Xu Z, Hall TL, Mycek M-A, Fowlkes JB and Cain CA, "Evolution of bubble

clouds produced in pulsed cavitational ultrasound therapy - histotripsy," IEEE

Trans Ultrason Ferroelectr Freq Control, vol. 55, pp. 1122-32, 2008.

[9] E. Vlaisavljevich, A. Maxwell, M. Warnez, E. Johnsen, C. A. Cain, and Z. Xu,

"Histotripsy-induced cavitation cloud initiation thresholds in tissues of different

mechanical properties," IEEE Trans Ultrason Ferroelectr Freq Control, vol. 61,

no. 2, pp. 341-52, Feb 2014, doi: 10.1109/TUFFC.2014.6722618.

[10] E. Vlaisavljevich, A. Maxwell, L. Mancia, E. Johnsen, C. Cain, and Z. Xu,

"Visualizing the Histotripsy Process: Bubble Cloud-Cancer Cell Interactions in a

Tissue-Mimicking Environment," Ultrasound Med Biol, vol. 42, no. 10, pp. 2466-

77, Oct 2016, doi: 10.1016/j.ultrasmedbio.2016.05.018.

[11] V. E. Kim Y, Owens GE, Allen SP, Cain CA and Xu Z, "In vivo transcostal

histotripsy therapy without aberration correction," Phys Med Biol, vol. 59, pp.

2553-68, 2014.

[12] E. Vlaisavljevich et al., "Non-Invasive Liver Ablation Using Histotripsy: Preclinical

Safety Study in an In Vivo Porcine Model," Ultrasound Med Biol, vol. 43, no. 6,

pp. 1237-1251, Jun 2017, doi: 10.1016/j.ultrasmedbio.2017.01.016.

Page 190: Determining the Oncological and Immunological Effects of

153

[13] A. M. Lake, Z. Xu, J. E. Wilkinson, C. A. Cain, and W. W. Roberts, "Renal

ablation by histotripsy--does it spare the collecting system?," J Urol, vol. 179, no.

3, pp. 1150-4, Mar 2008, doi: 10.1016/j.juro.2007.10.033.

[14] E. Vlaisavljevich, Y. Kim, G. Owens, W. Roberts, C. Cain, and Z. Xu, "Effects of

tissue mechanical properties on susceptibility to histotripsy-induced tissue

damage," (in eng), Phys Med Biol, vol. 59, no. 2, pp. 253-70, Jan 20 2014, doi:

10.1088/0031-9155/59/2/253.

[15] A. R. Smolock et al., "Robotically Assisted Sonic Therapy as a Noninvasive

Nonthermal Ablation Modality: Proof of Concept in a Porcine Liver Model,"

Radiology, vol. 287, no. 2, pp. 485-493, May 2018, doi:

10.1148/radiol.2018171544.

[16] N. R. Styn, J. C. Wheat, T. L. Hall, and W. W. Roberts, "Histotripsy of VX-2 tumor

implanted in a renal rabbit model," (in eng), J Endourol, vol. 24, no. 7, pp. 1145-

50, Jul 2010, doi: 10.1089/end.2010.0123.

[17] E. Vlaisavljevich et al., "Image-Guided Non-Invasive Ultrasound Liver Ablation

using Histotripsy: Feasibility Study in an In Vivo Porcine Model. ," Ultrasound

Med Biol, 2013.

[18] S. X. Vidal Jove J, Vlaisavljevich E, Cannata J, Duryea A, Miller R, Lee F and

Ziemlewicz T, "Phase I Study of Safety and Efficacy of Hepatic Histotripsy:

Preliminary Results of First in Man Experience with Robotically-Assisted Sonic

Therapy," in International Society of Therapeutic Ultrasound, Barcelona, Spain,

2019.

Page 191: Determining the Oncological and Immunological Effects of

154

[19] S. Qu et al., "Non-thermal histotripsy tumor ablation promotes abscopal immune

responses that enhance cancer immunotherapy," Journal for ImmunoTherapy of

Cancer, vol. 8, no. 1, 2020.

[20] T. Worlikar et al., "Non-invasive liver cancer ablation using histotripsy in an in

vivo murine hepatocellular carcinoma (HCC) model," in Ultrasonics Symposium

(IUS), 2017 IEEE International, 2017: IEEE, pp. 1-1.

[21] T. F. Vandamme, "Use of rodents as models of human diseases," (in eng), J

Pharm Bioallied Sci, vol. 6, no. 1, pp. 2-9, 2014, doi: 10.4103/0975-7406.124301.

[22] J. J. Tentler et al., "Patient-derived tumour xenografts as models for oncology

drug development," (in eng), Nature reviews. Clinical oncology, vol. 9, no. 6, pp.

338-50, Apr 17 2012, doi: 10.1038/nrclinonc.2012.61.

[23] M. G. S. Consortium, "Initial sequencing and comparative analysis of the mouse

genome," Nature, vol. 420, no. 6915, p. 520, 2002.

[24] J. Rossant and C. McKerlie, "Mouse-based phenogenomics for modelling human

disease," Trends in molecular medicine, vol. 7, no. 11, pp. 502-507, 2001.

[25] K. Paigen, "A miracle enough: the power of mice," Nature medicine, vol. 1, no. 3,

p. 215, 1995.

[26] M. De Jong and T. Maina, "Of mice and humans: are they the same?—

Implications in cancer translational research," Journal of Nuclear Medicine, vol.

51, no. 4, pp. 501-504, 2010.

[27] T. Flisikowska, A. Kind, and A. Schnieke, "Pigs as models of human cancers,"

Theriogenology, vol. 86, no. 1, pp. 433-7, Jul 1 2016, doi:

10.1016/j.theriogenology.2016.04.058.

Page 192: Determining the Oncological and Immunological Effects of

155

[28] K. M. Schachtschneider et al., "The Oncopig Cancer Model: An Innovative Large

Animal Translational Oncology Platform," Front Oncol, vol. 7, p. 190, 2017, doi:

10.3389/fonc.2017.00190.

[29] L. B. Schook et al., "A Genetic Porcine Model of Cancer," PLoS One, vol. 10, no.

7, p. e0128864, 2015, doi: 10.1371/journal.pone.0128864.

[30] K. Lee, K. Farrell, and K. Uh, "Application of genome-editing systems to enhance

available pig resources for agriculture and biomedicine," (in eng), Reprod Fertil

Dev, vol. 32, no. 2, pp. 40-49, Jan 2019, doi: 10.1071/rd19273.

[31] S. S. Pinho, S. Carvalho, J. Cabral, C. A. Reis, and F. Gartner, "Canine tumors: a

spontaneous animal model of human carcinogenesis," Transl Res, vol. 159, no.

3, pp. 165-72, Mar 2012, doi: 10.1016/j.trsl.2011.11.005.

[32] J. D. Schiffman and M. Breen, "Comparative oncology: what dogs and other

species can teach us about humans with cancer," Philos Trans R Soc Lond B

Biol Sci, vol. 370, no. 1673, Jul 19 2015, doi: 10.1098/rstb.2014.0231.

[33] M. Paoloni et al., "Defining the Pharmacodynamic Profile and Therapeutic Index

of NHS-IL12 Immunocytokine in Dogs with Malignant Melanoma," PLoS One, vol.

10, no. 6, p. e0129954, 2015, doi: 10.1371/journal.pone.0129954.

[34] M. Paoloni et al., "Prospective molecular profiling of canine cancers provides a

clinically relevant comparative model for evaluating personalized medicine

(PMed) trials," PLoS One, vol. 9, no. 3, p. e90028, 2014, doi:

10.1371/journal.pone.0090028.

[35] M. Paoloni, S. Lana, D. Thamm, C. Mazcko, and S. Withrow, "The creation of the

Comparative Oncology Trials Consortium Pharmacodynamic Core: Infrastructure

Page 193: Determining the Oncological and Immunological Effects of

156

for a virtual laboratory," Vet J, vol. 185, no. 1, pp. 88-9, Jul 2010, doi:

10.1016/j.tvjl.2010.04.019.

[36] M. C. Paoloni et al., "Rapamycin pharmacokinetic and pharmacodynamic

relationships in osteosarcoma: a comparative oncology study in dogs," PLoS

One, vol. 5, no. 6, p. e11013, Jun 08 2010, doi: 10.1371/journal.pone.0011013.

[37] I. Gordon, M. Paoloni, C. Mazcko, and C. Khanna, "The Comparative Oncology

Trials Consortium: using spontaneously occurring cancers in dogs to inform the

cancer drug development pathway," PLoS Med, vol. 6, no. 10, p. e1000161, Oct

2009, doi: 10.1371/journal.pmed.1000161.

[38] M. C. Paoloni et al., "Launching a novel preclinical infrastructure: comparative

oncology trials consortium directed therapeutic targeting of TNFalpha to cancer

vasculature," PLoS One, vol. 4, no. 3, p. e4972, 2009, doi:

10.1371/journal.pone.0004972.

[39] C. A. London et al., "Preclinical evaluation of the novel, orally bioavailable

Selective Inhibitor of Nuclear Export (SINE) KPT-335 in spontaneous canine

cancer: results of a phase I study," PLoS One, vol. 9, no. 2, p. e87585, 2014, doi:

10.1371/journal.pone.0087585.

[40] J. E. Parsons, C. A. Cain, G. D. Abrams, and J. B. Fowlkes, "Pulsed cavitational

ultrasound therapy for controlled tissue homogenization," (in eng), Ultrasound in

Medicine and Biology, vol. 32, no. 1, pp. 115-29, Jan 2006, doi: S0301-

5629(05)00374-1 [pii] 10.1016/j.ultrasmedbio.2005.09.005.

[41] J. E. Parsons, C. A. Cain, and J. B. Fowlkes, "Cost-effective assembly of a basic

fiber-optic hydrophone for measurement of high-amplitude therapeutic ultrasound

Page 194: Determining the Oncological and Immunological Effects of

157

fields," J Acoust Soc Am, vol. 119, no. 3, pp. 1432-40, Mar 2006, doi:

10.1121/1.2166708.

[42] E. Vlaisavljevich, T. Gerhardson, T. Hall, and Z. Xu, "Effects of f-number on the

histotripsy intrinsic threshold and cavitation bubble cloud behavior," Phys Med

Biol, vol. 62, no. 4, pp. 1269-1290, Feb 21 2017, doi: 10.1088/1361-

6560/aa54c7.

[43] H. R. Holmes et al., "Focused ultrasound extraction (FUSE) for the rapid

extraction of DNA from tissue matrices," (in English), Methods Ecol Evol, Oct 20

2020, doi: 10.1111/2041-210x.13505.

[44] J. Khirallah et al., "Nanoparticle-mediated histotripsy (NMH) using

perfluorohexane 'nanocones'," Phys Med Biol, vol. 64, no. 12, p. 125018, Jun 20

2019, doi: 10.1088/1361-6560/ab207e.

[45] R. M. Brock et al., "Patient derived xenografts expand human primary pancreatic

tumor tissue availability for ex vivo irreversible electroporation testing," Frontiers

in Oncology, vol. 10, 2020.

[46] D. N. Kwon et al., "Production of biallelic CMP-Neu5Ac hydroxylase knock-out

pigs," (in eng), Sci Rep, vol. 3, p. 1981, 2013, doi: 10.1038/srep01981.

[47] J. T. Kang et al., "Production of GGTA1 targeted pigs using TALEN-mediated

genome editing technology," (in Eng), Journal of veterinary science, Aug 4 2015.

[48] B. P. Beaton et al., "Inclusion of homologous DNA in nuclease-mediated gene

targeting facilitates a higher incidence of bi-allelically modified cells," (in eng),

Xenotransplantation, vol. 22, no. 5, pp. 379-90, Sep-Oct 2015, doi:

10.1111/xen.12194.

Page 195: Determining the Oncological and Immunological Effects of

158

[49] K. Lee et al., "Engraftment of human iPS cells and allogeneic porcine cells into

pigs with inactivated RAG2 and accompanying severe combined

immunodeficiency," Proc Natl Acad Sci U S A, vol. 111, no. 20, pp. 7260-5, May

20 2014, doi: 10.1073/pnas.1406376111.

[50] S. Lei et al., "Increased and prolonged human norovirus infection in RAG2/IL2RG

deficient gnotobiotic pigs with severe combined immunodeficiency," Sci Rep, vol.

6, p. 25222, Apr 27 2016, doi: 10.1038/srep25222.

[51] Y. J. Choi et al., "Recombination activating gene-2(null) severe combined

immunodeficient pigs and mice engraft human induced pluripotent stem cells

differently," Oncotarget, vol. 8, no. 41, pp. 69398-69407, Sep 19 2017, doi:

10.18632/oncotarget.20626.

[52] J. T. Kang et al., "Generation of RUNX3 knockout pigs using CRISPR/Cas9-

mediated gene targeting," (in eng), Reproduction in domestic animals =

Zuchthygiene, vol. 51, no. 6, pp. 970-978, Dec 2016, doi: 10.1111/rda.12775.

[53] K. Lee et al., "Piglets produced from cloned blastocysts cultured in vitro with

GM‐CSF," Molecular reproduction and development, vol. 80, no. 2, pp. 145-154,

2013.

[54] L. Yuan, P. M. Jobst, and M. Weiss, "Gnotobiotic Pigs: From Establishing Facility

to Modeling Human Infectious Diseases," in Gnotobiotics: Elsevier, 2017, pp.

349-368.

[55] A. V. M. Association, "Animal Health Studies Database," ed, 2016.

[56] T. Worlikar et al., "Histotripsy for Non-Invasive Ablation of Hepatocellular

Carcinoma (HCC) Tumor in a Subcutaneous Xenograft Murine Model," in 2018

Page 196: Determining the Oncological and Immunological Effects of

159

40th Annual International Conference of the IEEE Engineering in Medicine and

Biology Society (EMBC), 2018: IEEE, pp. 6064-6067.

[57] S. Lei et al., "Increased and prolonged human norovirus infection in RAG2/IL2RG

deficient gnotobiotic pigs with severe combined immunodeficiency," Scientific

reports, vol. 6, p. 25222, 2016.

[58] K. Lee et al., "Engraftment of human iPS cells and allogeneic porcine cells into

pigs with inactivated RAG2 and accompanying severe combined

immunodeficiency," Proceedings of the National Academy of Sciences, vol. 111,

no. 20, pp. 7260-7265, 2014.

[59] M. T. Basel et al., "Human xenografts are not rejected in a naturally occurring

immunodeficient porcine line: a human tumor model in pigs," BioResearch open

access, vol. 1, no. 2, pp. 63-68, 2012.

[60] A. K. LeBlanc et al., "Creation of an NCI comparative brain tumor consortium:

informing the translation of new knowledge from canine to human brain tumor

patients," Neuro Oncol, vol. 18, no. 9, pp. 1209-18, Sep 2016, doi:

10.1093/neuonc/now051.

[61] VT-VMCVM. "VT-VMCVM Website." http://www.vetmed.vt.edu/clinical-trials/

(accessed.

[62] M. L. Hauck and M. L. Oblak, "19 - Tumors of the Skin and Subcutaneous

Tissues," in Withrow and MacEwen's Small Animal Clinical Oncology (Sixth

Edition), D. M. Vail, D. H. Thamm, and J. M. Liptak Eds. St. Louis (MO): W.B.

Saunders, 2020, pp. 352-366.

Page 197: Determining the Oncological and Immunological Effects of

160

[63] J. M. Liptak and N. I. Christensen, "22 - Soft Tissue Sarcomas," in Withrow and

MacEwen's Small Animal Clinical Oncology (Sixth Edition), D. M. Vail, D. H.

Thamm, and J. M. Liptak Eds. St. Louis (MO): W.B. Saunders, 2019, pp. 404-

431.

[64] J. C. Friebele, J. Peck, X. Pan, M. Abdel-Rasoul, and J. L. Mayerson,

"Osteosarcoma: A Meta-Analysis and Review of the Literature," Am J Orthop

(Belle Mead NJ), vol. 44, no. 12, pp. 547-53, Dec 2015. [Online]. Available:

https://www.ncbi.nlm.nih.gov/pubmed/26665241.

[65] J. M. Fenger, C. A. London, and W. C. Kisseberth, "Canine osteosarcoma: a

naturally occurring disease to inform pediatric oncology," ILAR J, vol. 55, no. 1,

pp. 69-85, 2014, doi: 10.1093/ilar/ilu009.

[66] S. J. Withrow, B. E. Powers, R. C. Straw, and R. M. Wilkins, "Comparative

aspects of osteosarcoma. Dog versus man," Clin Orthop Relat Res, no. 270, pp.

159-68, Sep 1991. [Online]. Available:

http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=

Citation&list_uids=1884536.

[67] M. Paoloni et al., "Canine tumor cross-species genomics uncovers targets linked

to osteosarcoma progression," BMC genomics, vol. 10, p. 625, 2009, doi:

10.1186/1471-2164-10-625.

[68] F. Mueller, B. Fuchs, and B. Kaser-Hotz, "Comparative biology of human and

canine osteosarcoma," Anticancer research, vol. 27, no. 1A, pp. 155-64, Jan-Feb

2007. [Online]. Available: http://www.ncbi.nlm.nih.gov/pubmed/17352227.

Page 198: Determining the Oncological and Immunological Effects of

161

[69] E. Morello, M. Martano, and P. Buracco, "Biology, diagnosis and treatment of

canine appendicular osteosarcoma: similarities and differences with human

osteosarcoma," Vet J, vol. 189, no. 3, pp. 268-77, Sep 2011, doi:

10.1016/j.tvjl.2010.08.014.

[70] K. S. Rankin, M. Starkey, J. Lunec, C. H. Gerrand, S. Murphy, and S. Biswas, "Of

dogs and men: comparative biology as a tool for the discovery of novel

biomarkers and drug development targets in osteosarcoma," Pediatr Blood

Cancer, vol. 58, no. 3, pp. 327-33, Mar 2012, doi: 10.1002/pbc.23341.

[71] R. Gorlick and C. Khanna, "Osteosarcoma," J Bone Miner Res, vol. 25, no. 4, pp.

683-91, Apr 2010, doi: 10.1002/jbmr.77.

[72] J. L. Rowell, D. O. McCarthy, and C. E. Alvarez, "Dog models of naturally

occurring cancer," Trends Mol Med, vol. 17, no. 7, pp. 380-8, Jul 2011, doi:

10.1016/j.molmed.2011.02.004.

[73] L. Mirabello, R. J. Troisi, and S. A. Savage, "International osteosarcoma

incidence patterns in children and adolescents, middle ages and elderly

persons," Int J Cancer, vol. 125, no. 1, pp. 229-34, Jul 01 2009, doi:

10.1002/ijc.24320.

[74] S. F. Altekruse, K. A. McGlynn, and M. E. Reichman, "Hepatocellular carcinoma

incidence, mortality, and survival trends in the United States from 1975 to 2005,"

Journal of clinical oncology : official journal of the American Society of Clinical

Oncology, vol. 27, no. 9, pp. 1485-91, Mar 20 2009, doi:

10.1200/JCO.2008.20.7753.

Page 199: Determining the Oncological and Immunological Effects of

162

[75] S. A. Shah, J. K. Smith, Y. Li, S. C. Ng, J. E. Carroll, and J. F. Tseng,

"Underutilization of therapy for hepatocellular carcinoma in the medicare

population," Cancer, vol. 117, no. 5, pp. 1019-26, Mar 01 2011, doi:

10.1002/cncr.25683.

[76] C. J. Sonnenday, J. B. Dimick, R. D. Schulick, and M. A. Choti, "Racial and

geographic disparities in the utilization of surgical therapy for hepatocellular

carcinoma," J Gastrointest Surg, vol. 11, no. 12, pp. 1636-46; discussion 1646,

Dec 2007, doi: 10.1007/s11605-007-0315-8.

[77] S. Chacko and S. Samanta, ""Hepatocellular carcinoma: A life-threatening

disease"," Biomed Pharmacother, vol. 84, pp. 1679-1688, Dec 2016, doi:

10.1016/j.biopha.2016.10.078.

[78] D. R. Strombeck, "Clinicopathologic features of primary and metastatic neoplastic

disease of the liver in dogs," J Am Vet Med Assoc, vol. 173, no. 3, pp. 267-9, Aug

01 1978. [Online]. Available: https://www.ncbi.nlm.nih.gov/pubmed/211107.

[79] J. M. Liptak et al., "Massive hepatocellular carcinoma in dogs: 48 cases (1992-

2002)," J Am Vet Med Assoc, vol. 225, no. 8, pp. 1225-30, Oct 15 2004. [Online].

Available: https://www.ncbi.nlm.nih.gov/pubmed/15521445.

[80] C. Weisse, C. A. Clifford, D. Holt, and J. A. Solomon, "Percutaneous arterial

embolization and chemoembolization for treatment of benign and malignant

tumors in three dogs and a goat," J Am Vet Med Assoc, vol. 221, no. 10, pp.

1430-6, 1419, Nov 15 2002. [Online]. Available:

https://www.ncbi.nlm.nih.gov/pubmed/12458612.

Page 200: Determining the Oncological and Immunological Effects of

163

[81] Y. K. Cho, H. Rhim, and S. Noh, "Radiofrequency ablation versus surgical

resection as primary treatment of hepatocellular carcinoma meeting the Milan

criteria: a systematic review," J Gastroenterol Hepatol, vol. 26, no. 9, pp. 1354-

60, Sep 2011, doi: 10.1111/j.1440-1746.2011.06812.x.

[82] R. Z. Swan, D. Sindram, J. B. Martinie, and D. A. Iannitti, "Operative microwave

ablation for hepatocellular carcinoma: complications, recurrence, and long-term

outcomes," J Gastrointest Surg, vol. 17, no. 4, pp. 719-29, Apr 2013, doi:

10.1007/s11605-013-2164-y.

[83] M. Zhang, H. Ma, J. Zhang, L. He, X. Ye, and X. Li, "Comparison of microwave

ablation and hepatic resection for hepatocellular carcinoma: a meta-analysis,"

Onco Targets Ther, vol. 10, pp. 4829-4839, 2017, doi: 10.2147/OTT.S141968.

[84] N. Lucchina et al., "Current role of microwave ablation in the treatment of small

hepatocellular carcinomas," Ann Gastroenterol, vol. 29, no. 4, pp. 460-465, Oct-

Dec 2016, doi: 10.20524/aog.2016.0066.

[85] P. Liang and Y. Wang, "Microwave ablation of hepatocellular carcinoma,"

Oncology, vol. 72 Suppl 1, pp. 124-31, 2007, doi: 10.1159/000111718.

[86] D. J. Niemeyer et al., "Optimal ablation volumes are achieved at submaximal

power settings in a 2.45-GHz microwave ablation system," Surg Innov, vol. 22,

no. 1, pp. 41-5, Feb 2015, doi: 10.1177/1553350614532535.

[87] F. Wu et al., "Extracorporeal high intensity focused ultrasound ablation in the

treatment of 1038 patients with solid carcinomas in China: an overview," Ultrason

Sonochem, vol. 11, no. 3-4, pp. 149-54, May 2004, doi:

10.1016/j.ultsonch.2004.01.011.

Page 201: Determining the Oncological and Immunological Effects of

164

[88] C. X. Li et al., "Analysis of clinical effect of high-intensity focused ultrasound on

liver cancer," World J Gastroenterol, vol. 10, no. 15, pp. 2201-4, Aug 1 2004, doi:

10.3748/wjg.v10.i15.2201.

[89] F. Wu et al., "Pathological changes in human malignant carcinoma treated with

high-intensity focused ultrasound," Ultrasound Med Biol, vol. 27, no. 8, pp. 1099-

106, Aug 2001, doi: 10.1016/s0301-5629(01)00389-1.

[90] B. R. Partridge et al., "High-Frequency Irreversible Electroporation for Treatment

of Primary Liver Cancer: A Proof-of-Principle Study in Canine Hepatocellular

Carcinoma," (in eng), J Vasc Interv Radiol, vol. 31, no. 3, pp. 482-491.e4, Mar

2020, doi: 10.1016/j.jvir.2019.10.015.

[91] N. Beitel-White, R. C. G. Martin, Y. Li, R. M. Brock, I. C. Allen, and R. V.

Davalos, "Real-time prediction of patient immune cell modulation during

irreversible electroporation therapy," (in eng), Sci Rep, vol. 9, no. 1, p. 17739,

Nov 28 2019, doi: 10.1038/s41598-019-53974-w.

[92] V. M. Ringel-Scaia et al., "High-frequency irreversible electroporation is an

effective tumor ablation strategy that induces immunologic cell death and

promotes systemic anti-tumor immunity," EBioMedicine, vol. 44, pp. 112-125, Jun

2019, doi: 10.1016/j.ebiom.2019.05.036.

[93] Z. M. Bamboat et al., "Human liver dendritic cells promote T cell

hyporesponsiveness," Journal of immunology, vol. 182, no. 4, pp. 1901-11, Feb

15 2009, doi: 10.4049/jimmunol.0803404.

Page 202: Determining the Oncological and Immunological Effects of

165

[94] A. P. Jewell, "Is the liver an important site for the development of immune

tolerance to tumours?," Med Hypotheses, vol. 64, no. 4, pp. 751-4, 2005, doi:

10.1016/j.mehy.2004.10.002.

[95] P. Guha, J. Reha, and S. C. Katz, "Immunosuppression in liver tumors: opening

the portal to effective immunotherapy," Cancer Gene Ther, vol. 24, no. 3, pp.

114-120, Mar 2017, doi: 10.1038/cgt.2016.54.

[96] I. A. Siddiqui et al., "Induction of rapid, reproducible hepatic ablations using next-

generation, high frequency irreversible electroporation (H-FIRE) in vivo," HPB :

the official journal of the International Hepato Pancreato Biliary Association, vol.

18, no. 9, pp. 726-34, Sep 2016, doi: 10.1016/j.hpb.2016.06.015.

[97] C. M. Goutal, B. L. Brugmann, and K. A. Ryan, "Insulinoma in dogs: a review," J

Am Anim Hosp Assoc, vol. 48, no. 3, pp. 151-63, May-Jun 2012, doi:

10.5326/JAAHA-MS-5745.

[98] T. Schermerhorn, "Canine insulinoma as a model for studying molecular genetics

of tumorigenesis and metastasis," Vet J, vol. 197, no. 2, pp. 126-7, Aug 2013,

doi: 10.1016/j.tvjl.2013.04.017.

[99] M. Gillard et al., "Naturally occurring melanomas in dogs as models for non-UV

pathways of human melanomas," Pigment Cell Melanoma Res, vol. 27, no. 1, pp.

90-102, Jan 2014, doi: 10.1111/pcmr.12170.

[100] B. Hernandez, H. A. Adissu, B. R. Wei, H. T. Michael, G. Merlino, and R. M.

Simpson, "Naturally Occurring Canine Melanoma as a Predictive Comparative

Oncology Model for Human Mucosal and Other Triple Wild-Type Melanomas," Int

J Mol Sci, vol. 19, no. 2, Jan 30 2018, doi: 10.3390/ijms19020394.

Page 203: Determining the Oncological and Immunological Effects of

166

[101] R. M. Simpson et al., "Sporadic naturally occurring melanoma in dogs as a

preclinical model for human melanoma," Pigment Cell Melanoma Res, vol. 27,

no. 1, pp. 37-47, Jan 2014, doi: 10.1111/pcmr.12185.

[102] M. M. Rahman et al., "Transcriptome analysis of dog oral melanoma and its

oncogenic analogy with human melanoma," Oncol Rep, vol. 43, no. 1, pp. 16-30,

Jan 2020, doi: 10.3892/or.2019.7391.

[103] A. Prouteau and C. Andre, "Canine Melanomas as Models for Human

Melanomas: Clinical, Histological, and Genetic Comparison," Genes (Basel), vol.

10, no. 7, Jun 30 2019, doi: 10.3390/genes10070501.

[104] W. Supsavhad, W. P. Dirksen, C. K. Martin, and T. J. Rosol, "Animal models of

head and neck squamous cell carcinoma," Vet J, vol. 210, pp. 7-16, Apr 2016,

doi: 10.1016/j.tvjl.2015.11.006.

[105] R. F. Hoyt and S. J. Withrow, "Oral malignancy in the dog.," Journal of the

American Hospital Association, vol. 20, pp. 83-92, 1984.

[106] R. J. Todoroff and R. S. Brodey, "Oral and pharyngeal neoplasia in the dog: a

retrospective survey of 361 cases," (in eng), J Am Vet Med Assoc, vol. 175, no.

6, pp. 567-71, Sep 15 1979. [Online]. Available:

http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=

Citation&list_uids=511751.

[107] J. K. Kosovsky, D. T. Matthiesen, S. M. Marretta, and A. K. Patnaik, "Results of

partial mandibulectomy for the treatment of oral tumors in 142 dogs," (in eng),

Vet Surg, vol. 20, no. 6, pp. 397-401, Nov-Dec 1991. [Online]. Available:

Page 204: Determining the Oncological and Immunological Effects of

167

http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=

Citation&list_uids=1369522.

[108] N. Farcas, B. Arzi, and F. J. Verstraete, "Oral and maxillofacial osteosarcoma in

dogs: a review," Vet Comp Oncol, vol. 12, no. 3, pp. 169-80, Sep 2014, doi:

10.1111/j.1476-5829.2012.00352.x.

[109] J. Rossmeisl, "Maximizing Local Access to Therapeutic Deliveries in

Glioblastoma. Part V: Clinically Relevant Model for Testing New Therapeutic

Approaches," in Glioblastoma, S. De Vleeschouwer Ed. Brisbane (AU): Codon

Publications, 2017.

[110] M. F. Lorenzo et al., "Temporal Characterization of Blood-Brain Barrier Disruption

with High-Frequency Electroporation," (in eng), Cancers (Basel), vol. 11, no. 12,

Nov 23 2019, doi: 10.3390/cancers11121850.

[111] S. Demaria et al., "Ionizing radiation inhibition of distant untreated tumors

(abscopal effect) is immune mediated," (in eng), Int J Radiat Oncol Biol Phys, vol.

58, no. 3, pp. 862-70, Mar 1 2004, doi: 10.1016/j.ijrobp.2003.09.012.

[112] E. Vlaisavljevich, A. Maxwell, M. Warnez, E. Johnsen, C. Cain, and Z. Xu,

"Histotripsy-induced cavitation cloud initiation thresholds in tissues of different

mechanical properties," (in eng), IEEE Transactions on Ultrasonics,

Ferroelectrics, and Frequency Control, vol. 61, no. 2, pp. 341-52, Feb 2014, doi:

10.1109/TUFFC.2014.6722618.

[113] E. Vlaisavljevich et al., "Effects of tissue stiffness, ultrasound frequency, and

pressure on histotripsy-induced cavitation bubble behavior," Phys Med Biol, vol.

60, no. 6, pp. 2271-92, Mar 21 2015, doi: 10.1088/0031-9155/60/6/2271.

Page 205: Determining the Oncological and Immunological Effects of

168

[114] E. Vlaisavljevich, Z. Xu, A. Arvidson, L. Jin, W. Roberts, and C. Cain, "Effects of

thermal preconditioning on tissue susceptibility to histotripsy," Ultrasound in

medicine & biology, vol. 41, no. 11, pp. 2938-2954, 2015.

[115] H. Y. Ge et al., "Correlation between ultrasound reflection intensity and tumor

ablation ratio of late-stage pancreatic carcinoma in HIFU therapy: dynamic

observation on ultrasound reflection intensity," (in eng), ScientificWorldJournal,

vol. 2013, p. 852874, 2013, doi: 10.1155/2013/852874.

[116] H. Y. Ge et al., "High-intensity focused ultrasound treatment of late-stage

pancreatic body carcinoma: optimal tumor depth for safe ablation," Ultrasound

Med Biol, vol. 40, no. 5, pp. 947-55, May 2014, doi:

10.1016/j.ultrasmedbio.2013.11.020.

[117] K. E. Mitchell et al., "Outcomes of Limb-Sparing Surgery Using Two Generations

of Metal Endoprosthesis in 45 Dogs With Distal Radial Osteosarcoma. A

Veterinary Society of Surgical Oncology Retrospective Study," Vet Surg, vol. 45,

no. 1, pp. 36-43, Jan 2016, doi: 10.1111/vsu.12423.

[118] A. K. LeBlanc and C. N. Mazcko, "Improving human cancer therapy through the

evaluation of pet dogs," (in eng), Nat Rev Cancer, vol. 20, no. 12, pp. 727-742,

Dec 2020, doi: 10.1038/s41568-020-0297-3.

Page 206: Determining the Oncological and Immunological Effects of

169

Chapter 5. Establishing a SCID-Like Porcine Model of Human Cancer for Novel Therapy Development with Pancreatic Adenocarcinoma and Irreversible

Electroporation

Alissa Hendricks-Wenger, Kenneth N. Aycock, Margaret A. Nagai-Singer, Sheryl Coutermarsh-Ott, Melvin F. Lorenzo, Jessica Gannon, Kyungjun Uh, Kayla Farrell,

Natalie Beitel-White, Rebecca M. Brock, Alexander Simon, Holly A. Morrison, Joanne Tuohy, Sherrie Clark-Deener, Eli Vlaisavljevich, Rafael V. Davalos, Kiho Lee, Irving C.

Allen

This chapter has been previously published and used here with permission from: Hendricks-Wenger et al., Establishing a SCID-Like Porcine Model of Human Cancer for

Novel Therapy Development with Pancreatic Adenocarcinoma and Irreversible Electroporation, Scientific Reports, 2020, Nature Research

Page 207: Determining the Oncological and Immunological Effects of

170

5.1 Abstract

New therapies to treat pancreatic cancer are direly needed. However, efficacious

interventions lack a strong preclinical model that can recapitulate patients’ anatomy and

physiology. Likewise, the availability of human primary malignant tissue for ex vivo

studies is limited. These are significant limitations in the biomedical device field. We

have developed RAG2/IL2RG deficient pigs using CRISPR/Cas9 as a novel, large

animal model for use in cancer xenograft studies of human pancreatic adenocarcinoma.

In this proof-of-concept study, these novel pigs were successfully generated using on-

demand genetic modifications in embryos, circumventing the need for breeding and

husbandry. Human Panc01 cells injected subcutaneously into the ears of RAG2/IL2RG

deficient pigs demonstrated 100% engraftment with growth rates similar to those

typically observed in mouse models. Histopathology revealed no immune cell infiltration

and tumor morphology was highly consistent with the mouse models. The electrical

properties and response to irreversible electroporation of the tumor tissue were found to

be similar to excised human pancreatic cancer tumors. The ample tumor tissue

produced enabled improved accuracy and modeling of the electrical properties of tumor

tissue. Together, this suggests that this model will be useful and capable of bridging the

gap of translating therapies from the bench to clinical application.

Page 208: Determining the Oncological and Immunological Effects of

171

5.2 Keywords

Pancreatic Cancer; CRISPR/Cas9; Pig; Tumor Ablation; IRE

Page 209: Determining the Oncological and Immunological Effects of

172

5.3 Introduction

Despite relatively low rates of occurrence, pancreatic cancer accounts for a

disproportionately high rate of cancer associated death. Surgery may cure patients with

Stage I disease; however, more than 75% of patients will present with borderline or

unresectable disease (Stages II-IV).1,2 Chemotherapy and radiation treatment protocols

result in limited responses and are rarely curative.3-6 Thus, new therapies are direly

needed. Over the last decade, tumor ablation strategies, such as radiofrequency,

microwave, and cryoablation have emerged as promising therapeutic options for

pancreatic cancer.7-9 Prior to human trials, the ablation modalities developed to date for

pancreatic cancer were originally developed using models based on data generated

from healthy tissues, rodent models, and occasionally spontaneous tumors in veterinary

patients. Unfortunately, there are major limitations with each of these approaches that

have hindered tumor ablation device development and limited data necessary for

accurate treatment planning in human patients. Thus, the lack of clinically and

physiologically relevant pre-clinical pancreatic cancer models continues to be a

significant limitation.

Murine models are the dominant in vivo model in the biomedical engineering and

cancer biology fields and are essential in acquiring fundamental data. Mice are easy to

house and handle, relatively inexpensive, share many genetic similarities to humans,

and there are many transgenic and knock-out lines available to study specific disease

mechanisms, proteins, and pathways.10-15 While these advantages make murine models

Page 210: Determining the Oncological and Immunological Effects of

173

appealing, the gap between mice and humans is wide enough to prevent many of the

devices and therapies developed solely in mouse models from being safe and effective

in human patients. There are important anatomical differences between humans and

mice. Humans are approximately 2,500 times larger than mice, which accounts for

differences in many biological aspects, including metabolic rates, heart rates, and life

expectancy.15-19 Looking at carcinogenesis, mice develop malignant tumors more

readily than humans, due to significant differences in DNA repair and telomerase

regulation, among others.20 In an attempt to bridge this gap, more researchers are using

patient-derived tumor xenograft (PDX) models for oncological studies. In these models,

human tumors are engrafted into immunocompromised mice, allowing the local

microenvironment of the human tumor to be recapitulated in the mouse.11,21-24 While the

PDX model is an impressive improvement to mouse models, the anatomical differences

between mice and humans still presents a barrier to clinical relevance.

Porcine models have demonstrated increasing clinical relevance due to their similar

size, anatomy, and physiology to humans. Their large size is ideal and practical for

imaging technology studies and clinical device development actical.25 Porcine models

have been established as clinically-relevant models for toxicology, surgical procedures,

and designing equipment and instruments.26-28 Anatomical differences are especially

critical for the development and testing of medical devices for surgical and ablation

procedures. For instance, the safety and efficacy of minimal and non-invasive ablation

techniques such as radiofrequency ablation, microwave ablation, cryoablation,

irreversible electroporation (IRE), and focused ultrasound (HIFU, histotripsy) are difficult

to test in mice due to these anatomical differences. As a result, these devices are often

Page 211: Determining the Oncological and Immunological Effects of

174

tested in healthy large animal models, which can provide important data on treatment

safety and energy delivery, but make it difficult to assess the effects of the therapies in

diseased tissues. Therefore, the development of large animal tumor models would be

beneficial for pre-clinical safety and efficacy studies. Overall, pigs as a model are good

at recapitulating human cancers due to their similar size, genome, metabolism,

telomerase activity, and organ micro- and macro-structures.29,30 However, unlike murine

models, existing porcine models are not well-established or reproducible from subject-

to-subject.

The availability of abundant, high quality tumor tissue is a major limitation in the

development of medical devices that require ex vivo modeling and validation. This is

especially true in pancreatic cancer research, where human specimens from patients

are typically highly limited and restricted to small biopsies. Likewise, while post-mortem

tissue is more available, the quality of the tissue is often not ideal. While mice are

excellent bio-incubators for human tissue and cell lines, the size of tumor tissue is often

limited due to the relatively small size of the animal. We have found this to also be a

significant limitation in the development of irreversible electroporation (IRE) based

therapeutic strategies. While healthy porcine tissues and mouse models have been

critical for therapeutic development, we have found the lack of appropriate pancreatic

cancer tissues to be a limitation. It has long been known that electrical conductivity

deviates from its physiological value once tissues are removed from the body, but most

tissues remain minimally changed within 1 hour of removal.31

Members of our research team co-invented IRE as a non-thermal tissue ablation

modality capable of treating solid tumors32. IRE involves delivering a series of low

Page 212: Determining the Oncological and Immunological Effects of

175

energy, unipolar electric pulses through electrodes inserted directly into the tumor. The

induced electric field distribution produces structural defects in the target cell membrane

that cause cell death. IRE has been successfully adapted for the treatment of tumors

considered, until now, inoperable due to their proximity to critical structures such as

blood vessels, nerves, and ducts.33-36

In an effort to circumvent the limitations related to animal models of pancreatic

cancer and further advance the optimization of IRE utilizing human pancreatic cancer

tissue, we developed an immunodeficient porcine model that has proven ideal for

human cell transplantation and xenograft studies.37-41 Utilizing CRISPR/Cas9, we have

previously generated pigs with targeted deletions of RAG2/IL2RG, and established that

these pigs are immunodeficient and thrive under our unique gnotobiotic housing

conditions.41 This prior study utilized these pigs to study the pathogenesis of a human

virus.41 Expanding upon these previous studies, here we deploy the RAG2/IL2RG

deficient pigs as a novel tool to study pancreatic cancer. For the present study, we

describe the original proof-of-concept studies, utilizing these pigs to propagate the

human Panc01 pancreatic cancer cell line for use in ex vivo IRE modeling. This strategy

generated an ample supply of high-quality pancreatic tumor tissue for electrical property

modeling and IRE assessments. We believe that this model is a promising step towards

studying human cancer treatments in a highly-relevant system with the potential for

significant clinical impact.

Page 213: Determining the Oncological and Immunological Effects of

176

5.4 Materials And Methods

5.4.1 Generation of Immunodeficient Pigs

Table 5-1 Target sequences of RAG2 and IL2RG sgRNAs.

Target gene Sequence (PAM)

RAG2 5’ - GAATGACCATATCTGCCTTC(AGG)

5’ - GTATAGTCGAGGGAAAAGTA(TGG)

IL2RG

5’ - CCAACCTAACTCTGCACTAC(TGG)

5’ - TGGAAACGTTGAGAGTCCC(AGG)

5’ - AAACGTTGAGAGTCCCAGG(GGG)

Oocytes aspirated from sow ovaries were matured in maturation medium (TCM-199

medium supplemented with 0.5 IU ml−1 FSH, 0.5 IU ml−1 LH, 0.82 mM cysteine, 3.02 mM

glucose, 0.91 mM sodium pyruvate and 10 ng ml−1 EGF) for 40 – 44 hours. After in vitro

maturation, cumulus cells were removed by vortexing in the presence of hyaluronidase,

then mature oocytes extruding the first polar body were transferred to fertilization media

(modified Tris-buffered medium supplemented with 113.1 mM NaCl, 3 mM KCl, 7.5 mM

CaCl2, 11 mM glucose, 20 mM Tris, 2 mM caffeine, 5 mM sodium pyruvate and

2 mg ml−1 BSA). Extended semen was washed with PBS, then introduced into the IVF

dishes containing oocytes. The gametes were co-incubated for 5 hours at 38.5 °C and

5% CO2. Microinjection was conducted in manipulation media (TCM199 with 0.6 mM

NaHCO3, 2.9 mM HEPES, 30 mM NaCl, 10 ng ml−1 gentamicin and 3 mg ml−1 BSA)

covered with mineral oil on the heated stage of a Nikon inverted microscope (Nikon

Page 214: Determining the Oncological and Immunological Effects of

177

Corporation, Tokyo, Japan). Two sgRNAs targeting the RAG2 gene, three sgRNAs

targeting the IL2RG gene, and Cas9 mRNA (10 ng/µl sgRNA each and 20 ng/μl Cas9

mRNA) were introduced into the cytoplasm of the presumable zygotes using a FemtoJet

microinjector (Eppendorf, Hamburg, Germany). Target sequences of the sgRNAs can

be found in Table 1. The injected embryos were moved to culture media supplemented

with 10 ng ml−1 GM-CSF and incubated at 38.5 °C, 5% CO2, and 5% O2 until embryo

transfer 57. The cultured embryos were transferred into a surrogate gilt at day 4 post-IVF

by surgically transferring into the oviduct. Pregnancy was determined by ultrasound at

around day 30 of gestation. To genotype the mutations generated by CRISPR/Cas9

system, genomic DNA was isolated from tail-tips of the newborn piglets using PureLink

Genomic DNA kit (Thermo Fisher Scientific) following the manufacturer’s instructions.

Primers flanking projected double strand break (DSB) sites were designed to amplify

target region of RAG2 and IL2RG genes (Table 2). The target regions were PCR

amplified using Dream Taq DNA Polymerase (Thermo Fisher Scientific). PCR

conditions were as follows: initial denature at 95 °C for 2 min; denature at 95 °C for

30 sec, annealing at 60 °C for 30 sec, and extension at 72 °C for 30 sec for 34 cycles;

72 °C for 5 min; and holding at 4 °C. The PCR amplicons were sequenced to determine

the mutation types generated by the CRISPR/Cas9 system. Immunocompromised state

was verified with flow cytometry to analyze the presence B (CD79A), T (CD3E), and NK

(CD3E-/CD56+) cells.

Page 215: Determining the Oncological and Immunological Effects of

178

Table 5-2 Primers used to amplify sgRNA target regions.

Primer Sequence

RAG2_Forward GGCTTTCCCATAACCTGGATATTGGGTC

RAG2 Reverse CGTCTCAGACTCATCTTCCTCATCATCTT

IL2RG_Forward CGGTAATAATCATGACTAGAGGGAATGAAAGATTGATTTATC

IL2RG_Reverse GAGAGAAAGTTGGGTGTCTATAAAAGAAGGGAGAATTAAAACTG

5.4.2 Animal Care

Six pigs (ntotal=5 males and ntotal=1 female; n=2 male and n=1 female used for the

current study) were delivered through a sterile hysterectomy process previously

described by a board-certified large animal veterinarian.58 Pigs were monitored regularly

until fully recovered from anesthesia and able to eat independently. Tail snips were

taken from each piglet while still under the influence of anesthesia for genotyping, and

ears were clipped for numbering. The pigs were born into sterile, gnotobiotic isolators

and were fed sterile boxed milk throughout the study in order to prevent infections.58 All

animal experiments were approved and carried out in accordance with the Virginia Tech

Institutional Animal Care and Use Committee under IACUC protocol: 19-117-CVM.

5.4.3 Tumor Injection and Monitoring

Human Panc01 cells (National Cancer Institute DTP, DCTP Tumor Repository)

were grown up in DMEM supplemented with 10% FBS and 1% penicillin/ streptomycin

and removed from plates with Trypsin in EDTA. While the pigs were still under the effect

Page 216: Determining the Oncological and Immunological Effects of

179

of anesthesia from the hysterectomy, 1.2x106 Panc01 cells in 100 µL of Matrigel

(Corning, USA) were injected subcutaneously behind the ears of three pigs, with an

additional 100 µL of Matrigel behind the left ear. Tumors were measured three times per

week with plastic Vernier calipers. Diameter was calculated as the square root of the

product of the widest diameter and the diameter perpendicular to that. At the same point

of tumor measurement, weight and other health monitoring was also completed.

Following euthanasia, animals were necropsied and tissues were fixed in 10% formalin

for at least 24 hours. Paraffin-embedded formalin-fixed tissues were H&E stained and

evaluated by board-certified veterinary pathologist.

5.4.4 Immunohistochemistry

Pan02, PDX, and Panc01 samples (n=3/group) were subject to IHC-P staining of

CK19 (Thermo Fisher, MA5-12663). Paraffin-embedded formalin-fixed tissue sections

were deparaffinized following established xylene:ethonal wash protocol.59 Antigen

retrieval was achieved through 1-hour incubation at 95oC on a hot plate in sodium

citrate buffer with pH6 based upon the antibody manufacture’s suggestion. Once

cooled, slides were washed twice in tris-buffered saline (TBS) plus 0.025% Triton X-100

with gentle agitation. Primary antibody was diluted (1:250) in TBS with 1% BSA and

10% FBS overnight at 4 oC. Slides were washed four times in tris-buffered saline (TBS)

plus 0.025% Triton X-100 with gentle agitation, prior to 2 hours room temperature

incubation with secondary antibody (Cell Signaling, 7076P2) diluted (1:2000) in TBS

with 1% BSA and 10% FBS. Slides were washed three times for 5 minutes in TBS, then

Page 217: Determining the Oncological and Immunological Effects of

180

developed with DAB substrate (Abcam, ab64238) following manufactures guidelines

and counterstained with eosin and methyl blue.

5.4.5 Ultrasound Imaging

Animals were anesthetized with Xylazine/Telazol. Prior to imaging, pigs were

cleaned and dried, and had the area of interest shaved to remove hair that could

interfere with imaging. Ultrasound imaging was done using both linear and phased array

ultrasound transducers (L18-10L30H-4 and P8-3L0S1-6, TELEMED, Lithuania, EU).

Images were acquired on a laptop computer and saved for future analysis. Targets were

confirmed by a board-certified veterinarian.

5.4.6 Tissue Handling and Electroporation

Small rectangular sections of healthy or malignant tissue were excised, washed to

prevent blood coagulation, and placed in modified PBS to help maintain osmotic

pressure balance as previously described.60 Immediately prior to pulsing, a sample of

the tissue was removed from PBS, sectioned into smaller pieces, and placed into an

insulating cylindrical polydimethylsiloxane (PDMS) mold as previously described.61 The

mold had radius of 3 mm and thickness of 0.56 cm. The tissue-containing mold was

placed between a set of parallel plate electrodes connected to a pulse generator (BTX

ECM 830, Harvard Apparatus, Holliston, MA). Baseline electrical conductivity was

measured by delivering a low voltage (~20 V) potential across the electrodes. Electrical

impedance was then recorded from 1 to 106 Hz.

Page 218: Determining the Oncological and Immunological Effects of

181

All excised tumor and healthy tissues were analyzed within 1 hour of resection. Prior

to data analysis, all DC offsets were removed and a Butterworth filter was applied to

smooth the data and allow for automated analysis in MATLAB (MathWorks Inc., Natick,

MA, USA). To assess differences in electrical properties between our RAG2/IL2RG pig

Panc01 tumor model and primary human pancreatic cancer specimen, we compared

the initial tissue conductivities and the changes in tissue conductivity at 2.5 kV/cm.

Baseline electrical properties were recorded for healthy pancreas, prostate and brain

tissue excised from RAG2/IL2RG pigs. Prior to IRE delivery, a low voltage (~20 V) pulse

was administered to record sample-specific initial conductivity. This low potential pulse

does not cause electroporation-induced changes in electrical properties, and is thus

only diagnostic. The resulting value was used as a baseline to determine percent

differences in tissue conductivity during the subsequent high voltage, 100-pulse IRE

protocol. An electric field strength of either 500, 750, 1000, 1500, 2000, 2500, and 3,000

V/cm was delivered to each sample (n=1 for each electric field strength, n=7 total

samples). For comparison, the data acquired in this study are reported alongside tissue

conductivity data from primary human pancreatic cancer, as previously reported in

Beitel-White et al., primary pancreatic ductal adenocarcinoma (PDAC) tissue

propagated in mice from Brock et al., as well as, data obtained from literature for

healthy pancreas, prostate, and brain tissue.24,46,62-68

To monitor changes in temperature due to joule heating, a general-purpose STB

fiber optic thermometry probe (LumaSense, Santa Clara, CA, USA) was inserted at the

midpoint of the cylindrical tissue sample. Temperature was recorded at a frequency of 2

Hz using a Luxtron m3300 Biomedical Lab Kit (LumaSense, Santa Clara, CA, USA). An

Page 219: Determining the Oncological and Immunological Effects of

182

unpaired t-test with Welch’s correction was used to compare the mean initial

conductivity.

Figure 5-1 Generation of RAG/ IL2RG knockout pigs. (A) Schematic approach for RAG2/IL2RG knockout pig production. First, the sgRNA and Cas9 mRNA was synthesized via in vitro transcription. Second, an optimal concentration of sgRNA and Cas9 was injected into presumable zygotes. Third, the injected embryos were transferred into a surrogate gilt. At the end of gestation, piglets were born via hysterectomy and maintained in gnotobiotic isolators. (B) Summary of piglet genotypes in IL2RG and RAG2 gene. Six piglets were born and analyzed. During PCR analysis, RAG2 was not amplified from the Piglet #2 DNA sample, suggesting a large deletion. Biallelic genotype indicates two differently modified alleles. Homozygous indicates one type of modified alleles. (C) Genotype of Piglet 1 as a representation of small deletions or insertions introduced by the CRISPR/Cas9 system.

Page 220: Determining the Oncological and Immunological Effects of

183

5.5 Results

5.5.1 Generation of immunodeficient pigs carrying targeted deletions in RAG2/IL2RG

using the CRISPR/Cas9 system

Previous studies have utilized immunodeficient pigs in infectious disease studies

and to evaluate engraftment of human iPS cells.42-44 However, these pigs have yet to be

widely utilized in cancer research models. CRISPR/Cas9 RNAs were introduced into the

cytoplasm of in vitro fertilized pig oocytes (Fig. 1A). To disrupt pig immune system, the

RAG2 and IL2RG genes were targeted.42 In vitro fertilized oocytes (n = 454) were

injected with the targeted RNA for the CRISPR/Cas9 system, then cultured in vitro. On

day 4 post-IVF, 155 embryos at between the 8-cell and morula stage were transferred

to a surrogate gilt and subsequently six piglets were derived by hysterectomy. An

important advantage of this strategy is the ability to generate the RAG2/IL2RG deficient

pigs “on-demand”, without the need to maintain breeding colonies. Once born, the

immunocompromised state of the pigs requires them to be maintained in gnotobiotic

isolators under germ-free condition. Genotyping was conducted using genomic DNA

collected from tail-snips. Similar to our previous report,41 genotyping revealed that none

of the six piglets have wild-type sequences in either of the RAG2 or IL2RG genes (Fig.

1B detailed in Supplementary Fig. S1). All six piglets carried small insertion or deletion

mutations in both RAG2 and IL2RG genes (Fig. 1B). The genotype of piglet 1 is shown

as an example (Fig. 1C). A RAG2 fragment flanking the CRISPR/Cas9-induced DSBs

site could not be amplified from one piglet by PCR, suggesting a larger deletion caused

Page 221: Determining the Oncological and Immunological Effects of

184

by CRISPR/Cas9 system. For tumor engraftment studies, 3 of the 6 pigs were randomly

selected for subsequent Panc01 studies. Aligned with the RAG2/IL2RG knockout

establishing study,42 the piglets presented B-T-NK-SCID phenotype (Fig. 2).

Page 222: Determining the Oncological and Immunological Effects of

185

Figure 5-2 Confirmation of SCID-Like Phenotype. FACS analysis to detect the presence of lymphocytes in RAG2/IL2RG knockout piglets. Compared to wild-type control, SCID piglet possessed lower level of B (CD79A), T (CD3E), and NK (CD3E-/CD56+) cells.

5.5.2 Panc01 tumors successfully engrafted in all pigs carrying the targeted

RAG2/IL2RG deletion

Figure 5-3 Successful Engraftment of Panc01 Cells. (A) Tumor growth was measured three times per week for each pig. All animals developed palpable tumors that continued to grow over the course of the study. (B) Pig weight was monitored throughout the duration of the study and utilized as a surrogate for animal morbidity.

The purpose of the current study was to generate human tumors of sufficient size

and quality in our unique immunocompromised pigs for ex vivo assessments of IRE.

However, due to this being the first use of these pigs to propagate tumor tissue and the

potential for incomplete knockout of the immune system due to inefficiencies in the

CRISPR/Cas9 strategy, we first established engraftment proficiency and growth rates

for the Panc01 xenografts. Within 24 hours of the initial injection, tumor growth was

Page 223: Determining the Oncological and Immunological Effects of

186

observed for all Panc01 tumors. After an initial period of rapid growth, within 5 days of

xenograft, the rate of increase stabilized and steadily increased to the target diameter of

1.0 - 1.6cm by day 36 (Fig. 3A). No changes were observed in morbidity, mortality, or

clinical parameters associated with tumor progression demonstrated here by the

change in pig weight over time (Fig. 3B). All tumors were easily palpated and easily

observed by day 36 (Fig. 4A). The Matrigel control injection was not detected (Fig. 4B).

Additional assessments utilizing ultrasound imaging revealed clearly defined tumors,

with clear delineations from surrounding tissues (Fig. 4C). Necropsy revealed the

subcutaneous tumors remained within the dermal layer, and where easily palpated and

observable from underneath the dermal layers (Fig. 4D). Tumor measurements

collected extra-dermally, sub-dermally, and with ultrasound were comparable for each

tumor (Fig. 4A, C-E). At gross necropsy, no evidence of metastasis was identified in

draining or other lymph nodes, nor in organs with major vascular supply, such as the

spleen, liver, or lung.

Page 224: Determining the Oncological and Immunological Effects of

187

Figure 5-4 Subcutaneous Tumors Generated Ample, High-Quality Pancreatic Cancer Tissue for Subsequent Ex Vivo Tumor Ablation Assessments. (A) Tumor growth was readily observable above the skin behind the ear where the Panc01 cells were injected. (B) No observable foci were located in regions where Matrigel alone (control) was injected. (C) Ultrasound image confirmed the volumetric growth of the tumors. The tumor is outlined here in white for clarity. (D-E) Necropsy of the tumors confirmed that (D) they were confined to the dermis and (E) approximately the same size as measured extra-dermally.

5.5.3 Panc01 tumors are histopathologically similar to Pan02 and PDX tumors

generated in mice

Previous studies have utilized human xenografts in immunocompromised mice or

murine tumor injection into immunocompetent animals to generate tissue for ex vivo IRE

studies. However, the small size of the mice is a restriction for tumor growth and

limitation for any ultimate orthotopic studies. Due to the historic use of mice in IRE and

other tumor ablation modality development, we next compared the Panc01 tumors from

the RAG2/IL2RG pigs with Pan02 tumors generated in immunocompetent mice. In

Page 225: Determining the Oncological and Immunological Effects of

188

Figure 5-5 Histopathology Comparison of Pre-Clinical Pancreatic Cancer Animal Models. Human PDX, mouse Pan02, and human Panc01 were compared. (A) Subcutaneous tumors in the mouse Pan02 model are densely cellular often with central areas of necrosis (asterisk). Individual cells are spindle-shaped and arranged in vague, irregular streams. (B) Subcutaneous tumors in the PDX mice are also highly cellular and also exhibit foci of necrosis (asterisk). At higher magnification, these cells are more polygonal and are often arranged to form irregular tubular structures with lumens containing inflammatory cells and debris (arrows). (C) Subcutaneous tumors in the porcine Panc01 model are also densely cellular and variably exhibit foci of necrosis. Individual cells are more polygonal in shape and rarely attempt to recapitulate glandular structures (arrows).

Page 226: Determining the Oncological and Immunological Effects of

189

Figure 5-6 Immunohistochemical Confirmation of Ductal Cells in Pre-Clinical Pancreatic Cancer Animal Models. (A) Human PDX, (B) mouse Pan02, and (C) human Panc01 were compared for CK19 expression. Brown color indicates positive color. Nuclei are stained in dark blue, and remaining cell features are highlighted in light blue.

Page 227: Determining the Oncological and Immunological Effects of

190

general, cell line tumors from both animal models were highly similar. However, as

expected simply due to the immune system status of each animal, we did observe some

minor differences in tumor histopathology. In the murine Pan02 and in the murine PDX

subcutaneous pancreatic adenocarcinoma models, nodular tumors develop at the site

of injection (Fig. 5A and 5B, respectively). The Pan02 tumors are densely cellular with

minimal fibrovascular stroma and often variably-sized foci of necrosis develop centrally

within the tumor. Tumor cells are spindle in shape and tend to be arranged in vague,

indistinct streams. Similar to most malignant tumor cells of any origin, they often exhibit

marked pleomorphism (differences in cell and nuclear size and shape across the

population) with numerous mitotic figures scattered throughout. In the PDX model, cells

derived from human tumors are injected into immunodeficient NOD-Scid gamma (NSG)

mice. Microscopically, the tumors in these mice look different from those in the Pan02

model. For example, in the PDX model, tumor cells are more polygonal in shape

(characteristic of epithelial cells) and are often arranged into disorganized glandular

structures. In the RAG2/IL2RG pig xenografts, microscopically the tumors are

characterized by dense cellularity on minimal fibrovascular stroma, as seen in other

models (Fig. 5C). Tumor cells, resemble an intermediate between the two murine

models. Cells are more polygonal in shape and do rarely attempt to recapitulate

glandular structures. These are often indistinct and do not often exhibit lumens.

Localization of CK 19, an established ductal cell marker, was also analyzed in Pan02,

PDX, and Pan01 tumors in order to establish the ductal characteristics of the tissues.45

Murine Pan02 sections were not found to express CK19 (Fig. 6A). In the murine PDX

Page 228: Determining the Oncological and Immunological Effects of

191

and RAG2/IL2RG pig xenografts, CK19 expression was found throughout the tissues,

localizing in glandular and glandular-like structures respectively (Fig. 6B-C).

5.5.4 Panc01 tumors demonstrate similar electrical properties to primary patient derived

pancreatic cancer tissue and healthy porcine tissues

Figure 5-7 Primary human, SCID-like Panc01, and murine PDX tumors demonstrate similar conductivity increases following high voltage pulsed electric field. Ex vivo tissue samples directly from patients, RAG2/IL2RG animals, or NSG mice were sectioned and fit within a PDMS mold to impose a cylindrical shape factor. After sample preparation, individual samples were exposed to IRE pulses with varying electric field magnitudes. A) Summary of initial conductivities calculated from normal and cancerous SCID-like (black), human (red), and murine PDX (gray) tissue. B) The conductivity for each tissue type increased with the applied electric field. C) Percent increase in tissue conductivity at varying fields was determined from the sample-specific pre-pulse values. D) The adjusted conductivity is calculated from the percent difference values; here, the conductivities are normalized to the average initial tissue conductivity and adjusted based on the percent change.

Page 229: Determining the Oncological and Immunological Effects of

192

Figure 5-8 Impedance for healthy tissue excised from the RAG2/IL2RG pigs correlate adult swine and humans. A Gamry Reference 600 potentiostat (Gamry, Warminster, PA, US) was used to record tissue impedance within a frequency range of 103 to 106 Hz. The real part of the tissue impedance was used in the calculation of tissue conductivity for further comparison to values previously reported in literature. A) Comparison of electrical conductivity from SCID-like pig tissue to healthy porcine (pancreas/brain) or human (prostate) tissue. The B) pancreatic, C) brain, and D) prostate tissue demonstrate similar electrical behavior to their normal counterparts across a wide frequency range. Each data point (dot) represents reported values for normal tissue conductivity, while the black line represents data gathered in this study and the red line represents normal porcine tissue. (*p < 0.05).

There are several significant advantages of the Panc01 porcine xenograft model

over other currently existing models and even primary human pancreatic cancer tissue.

Specifically, the Panc01 tumors are readily available, high quality, and in sufficient

Page 230: Determining the Oncological and Immunological Effects of

193

volume for robust ex vivo studies. Thus, we next compared the electrical properties of

the Panc01 tissue with primary human pancreatic cancer tissue (PHPC) and murine-

propagated primary pancreatic ductal adenocarcinoma (PDAC) tissue as originally

reported in Brock et al. (Fig 7).24 The PHPC tissue (n = 7), excised Panc01 tumor

tissue (n = 7), and murine PDX tissue (n = 9) exhibited initial conductivities of 0.231

0.058 S/m, 0.242 0.043 S/m, and 0.225 0.070 S/m, respectively (Fig. 7A). No

statistically significant differences were found between these values. With an applied

electric field of 2.5 kV/cm, the mean intra-pulse conductivity increased to 0.288 S/m,

0.331 S/m, and 0.386 S/m for the PHPC, Panc01, and murine PDX tissue, respectively

(Fig. 7B). The relationship between the treated tumors is more clearly seen as a percent

increase from base line, with corresponding adjusted conductivity (Fig. 7C-D).

Directly comparing the healthy porcine pancreas in RAG2/IL2RG pigs to human

pancreas, the initial conductivity was 0.133 0.0059 S/m and 0.197 0.103 S/m (p =

0.0678), respectively (Fig. 7A). In addition to the healthy pig pancreas, we also

compared results with healthy brain and prostate. The EIS results for healthy brain,

pancreas, and prostate are reported as low frequency (10 kHz) and high frequency (1

MHz) measurements for direct comparison to literature values; the impedance spectrum

for each tissue and the comparison to literature values are shown in Figure 8. At low

frequency, the conductivity values for healthy tissue analyzed from the RAG2/IL2RG

model for brain (n = 8), pancreas (n = 3), and prostate (n = 7) tissue measured at 0.175

0.017 S/m, 0.145 0.005 S/m, and 0.343 0.039 S/m. In this same frequency, the

conductivity data measured from the standard healthy porcine model for brain (n = 5)

and pancreatic (n = 4) tissues measured at 0.167 0.016 S/m and 0.188 0.02 S/m,

Page 231: Determining the Oncological and Immunological Effects of

194

respectively (Fig. 8A). A t-test was used to determine statistically significant differences

between the RAG2/IL2RG pig healthy tissue and the standard porcine model healthy

tissue; no statistical differences were detected for brain tissue (p = 0.4401), but a

statistical difference was detected for the pancreatic tissue (p = 0.021). High frequency

conductivity values for healthy tissue analyzed from the RAG2/IL2RG pig model for

brain (n = 8), pancreas (n = 3), and prostate (n = 7) tissue measured 0.294 0.021 S/m,

0.371 0.009 S/m, and 0.508 0.043 S/m (Fig. 8A). In this same frequency, conductivity

of the standard healthy porcine model for brain (n = 5) and pancreatic (n = 4) tissue

measured at 0.276 0.016 S/m and 0.383 0.009 S/m, respectively. Due to the lack of

easily accessible, normal porcine prostatic tissue, we compared SCID-like prostate

conductivity to that collected from freshly excised human prostates and reported in

Halter et. al, 2007.46 Halter and colleagues measured low- and high-frequency

conductivities of 0.43 0.27 S/m and 0.60 0.32 S/m, respectively, neither of which are

significantly different (p = 0.514 and p = 0.551) than the values reported herein for

SCID-like prostate conductivity (Fig. 8A). A t-test was used to determine statistically

significant differences between the RAG2/IL2RG healthy tissue and the standard

porcine model healthy tissue; no statistical differences were detected for brain (p =

0.1092) or pancreatic tissue (p = 0.1349) across porcine models. Due to the lack of

healthy porcine prostate data available, no direct comparisons were made between the

SCID-like prostate and conventional porcine prostate tissue at the abovementioned

frequencies.

Page 232: Determining the Oncological and Immunological Effects of

195

5.6 Discussion

With their larger size compared to rodents and controlled immune system

knockdown, RAG2/IL2RG pigs created with the CRISPR/Cas9 system may be used as

a novel oncology model for translating developing therapies. They offer the opportunity

to grow xenograft tumors from immortalized cell lines, as well as, primary tumors from

patients, in an organism that can grow to a scale comparable to the size of humans.

Previous animal models have been limited due to small animal size, preventing the

development of tumors that are comparable in size and location to humans, and other

large animal models are less predictable and reproducible. With this controlled

immunocompromised model, the capability to engraft tumors would allow for more

relevant and efficient pre-clinical trials.

The growth of the tumors in the subcutaneous space in the pigs was notable

immediately after engraftment and maintained a relatively steady growth rate beyond

that point. This controlled growth in conjunction with histology, which showed no signs

of host immune rejection from the myeloid cells that remain in immunocompromised

conditions, indicate that this model is capable of supporting xenografts even with the

moderate cell injections of 1.2x106 used here. The current study stopped tumor growth

at 36 days as a pre-determined end point for consistency and direct comparisons with

the mouse models, not due to health effects or tumor burden, which commonly prevent

murine models from producing larger tumors. Beyond that, should this model be used

for subcutaneous treatments, the immunocompromised state of the pigs did not leave

them susceptible to increased systemic disease, as seen by their steady increase in

Page 233: Determining the Oncological and Immunological Effects of

196

weight, lack of clinical symptoms, and their lack of metastases found on necropsy.

Future studies should be conducted in order to determine if orthotopic engraftment can

be supported by the animals without notable health effects.

Comparison of the porcine xenografted Panc01 tumors to the murine Pan02

tumors and PDAC murine xenografted PDX tumors, is based upon the latter two

model’s well established roll in studying cancer therapies.24,47,48 Based on histology, the

xenograft tumors also share comparable features of interest with accepted human-

tumor xenograft models. The PDX model is similar to the Pan02 model in that tumor

cells are subcutaneously injected into the mouse; however, the model differs in several

important ways (Fig 5A-B). The PDX tumors are grown in NSG, immunocompromised,

mice, and develop glandular structures that express CK19. These glands often have

distinct lumens and will often be filled with clear space, small amounts of secretory

product, inflammatory cells, and/or cellular debris. The most common type of pancreatic

tumor in humans is pancreatic ductal adenocarcinoma, which is histologically

characterized by polygonal cells often arranged into glandular structures. Similar to the

PDX models, the Panc01 tumors from the immunocompromised pigs also closely

resemble this microscopic morphology and are consistent with observations in human

patients. This includes cells with marked pleomorphism and prominent mitotic figures.

Another feature of the tumors in PDX mice is the fairly prominent fibrovascular stroma,

which is not present in the Pan02 tumor. However, the pig tumors do exhibit an

intermediate stroma again suggesting it is more similar to the PDX than the Pan02. The

tumor stroma seems to be a pretty important player and is often a barrier to treatment.49-

51 Additionally, agreeing with previous studies, we did not find ductal cell maker CK19

Page 234: Determining the Oncological and Immunological Effects of

197

on our Pan02 tumors.52 On the other hand, the murine PDX and porcine Panc01 tumors

both express this marker, establishing their similarity to PDAC tumors.53 Thus, this

model recapitulates many features found in patient adenocarcinoma and is comparable

to PDX tumors, which are currently considered superior animal model for tumor

propagation and ex vivo studies.

For the feasibility studies described here, we used IRE as an example

technology that can benefit from tumors propagated in the RAG2/IL2RG pigs. Due to

electroporation-based therapies relying on exposing targeted cells to a specific electric

field, the conductivities of the tissues in the region of interest are critical to determining

treatment outcome. Thus, conductivity is one of the principal parameters affecting the

production of lesions due to irreversible electroporation, and preclinical animal models

must exhibit similar electrical signatures to their human counterparts to maximize

clinical translatability. Additionally, the morphology of cells affects the local electric

potential they experience when an external electric field is applied.54-56 Thus, faithful

recapitulations of human tumor tissue should be similar in terms of cell shape, size,

distribution, and intracellular content as this will ultimately affect the treatment outcome.

Notably, the PDX tumors presented in this study have been shown to recapitulate the

histological characteristics of human PDAC samples.24 Thus, the similar morphology of

cells in the Panc01 tumors introduced here indicates similarity to primary tumor samples

as well. Though the Panc01 tumors exhibited fewer ductal structures than the human

PDX tissue, this did not introduce differences in measured electrical properties. This

similarity is likely the result of the similar cell size and packing density between the PDX

and Panc01 tissue (Fig. 5). However, it is possible that the relatively large samples

Page 235: Determining the Oncological and Immunological Effects of

198

(~0.2 cm3), excision process, and PBS wash introduce minute changes in tissue

structure/composition that limit the ability of our methods to resolve small spatial

differences in conductivity due to the presence of these ductal structures. Despite this,

we have shown that the bulk tissue response is similar between tissue models,

suggesting that the Panc01 platform exhibits a similar bulk response to applied fields as

currently established tissue surrogates.

To assess the electrical similarity of the subcutaneous Panc01 tumors to locally

advanced pancreatic cancer (LAPC) observed in humans, we conducted a series of

experiments in which electrical pulses were delivered across flat-plate electrodes to

determine electrical conductivity as a function of applied electric field. Our results

demonstrate that the Panc01 tumors grown in RAG2/IL2RG pigs have similar electrical

behavior to spontaneous tumors in humans, as well as murine-propagated human

PDAC tissue, increasing our confidence that the local electric field produced in the pig-

derived tumors will be similar to that which arises in human neoplasms.

Together, these data support the feasibility of the immunocompromised pig model

for evaluating the clinical relevance of novel medical devices and other surgical

procedures, including those that rely on accurate recapitulation of human tumor

morphology and electrical properties. Additionally, the anatomical features, including

size, of the RAG2/IL2RG pigs is more similar to that of humans compared to most

common pre-clinical animal models, making these swine ideal for prototype testing

without the need for a significant scale down. Here we have shown for the first time that

the dielectric properties of RAG2/IL2RG pigs are similar to those of humans. Overall,

these data indicate that this immunocompromised model is capable of recapitulating

Page 236: Determining the Oncological and Immunological Effects of

199

physiologically relevant tumors comparable to other commonly utilized tumor models

with several novel advantages. Together, this suggests that this model will be highly

useful and capable of bridging the gap of translating ablation therapies from the bench

to clinical application.

5.7 Abbreviations

SCID: Severe combined immunodeficiency; IRE: irreversible electroporation; PDX:

patient derived xenograft

5.8 Funding Sources

This work was supported by the Virginia Maryland College of Veterinary Medicine

(I.C.A.); The Virginia Tech Institute for Critical Technology and Applied Sciences Center

for Engineered Health (I.C.A. and R.V.D.); The Virginia Biosciences Health Research

Corporation Catalyst (R.V.D.); The Focused Ultrasound Foundation (I.C.A.); The

National Institutes of Health R21OD027062 (K.L.), R21 R21EB028429 (I.C.A.),

R01CA213423 (R.V.D.), and P01CA207206 (R.V.D.). The content is solely the

responsibility of the authors and does not necessarily represent the official views of the

NIH or any other funding agency.

Page 237: Determining the Oncological and Immunological Effects of

200

5.9 References

1 Howlader N, N. A., Krapcho M, Garshell J, Miller D, Altekruse SF, Kosary CL, Yu

M, Ruhl J, Tatalovich Z,Mariotto A, Lewis DR, Chen HS, Feuer EJ, Cronin KA

(eds). SEER Cancer Statistics Review, 1975-2012, National Cancer Institute.

Bethesda, MD. http://seer.cancer.gov/csr/1975_2012/, based on November 2014

SEER data submission, posted to the SEER web site, April 2015.

2 Riall, T. S. et al. Pancreatic cancer in the general population: improvements in

survival over the last decade. Journal of gastrointestinal surgery 10, 1212-1224

(2006).

3 Oettle, H. et al. Adjuvant chemotherapy with gemcitabine vs observation in

patients undergoing curative-intent resection of pancreatic cancer: a randomized

controlled trial. Jama 297, 267-277 (2007).

4 Conroy, T. et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic

cancer. New England Journal of Medicine 364, 1817-1825 (2011).

5 Sultana, A. et al. Meta-analyses of chemotherapy for locally advanced and

metastatic pancreatic cancer. Journal of Clinical Oncology 25, 2607-2615 (2007).

6 Stocken, D. et al. Meta-analysis of randomised adjuvant therapy trials for

pancreatic cancer. British journal of cancer 92, 1372-1381 (2005).

7 Arcidiacono, P. G. et al. Feasibility and safety of EUS-guided cryothermal

ablation in patients with locally advanced pancreatic cancer. Gastrointestinal

endoscopy 76, 1142-1151 (2012).

Page 238: Determining the Oncological and Immunological Effects of

201

8 Carrafiello, G. et al. Microwave ablation of pancreatic head cancer: safety and

efficacy. Journal of Vascular and Interventional Radiology 24, 1513-1520 (2013).

9 Girelli, R. et al. Feasibility and safety of radiofrequency ablation for locally

advanced pancreatic cancer. British Journal of Surgery 97, 220-225 (2010).

10 Vandamme, T. F. Use of rodents as models of human diseases. J Pharm

Bioallied Sci 6, 2-9, doi:10.4103/0975-7406.124301 (2014).

11 Tentler, J. J. et al. Patient-derived tumour xenografts as models for oncology

drug development. Nature reviews. Clinical oncology 9, 338-350,

doi:10.1038/nrclinonc.2012.61 (2012).

12 Consortium, M. G. S. Initial sequencing and comparative analysis of the mouse

genome. Nature 420, 520 (2002).

13 Rossant, J. & McKerlie, C. Mouse-based phenogenomics for modelling human

disease. Trends in molecular medicine 7, 502-507 (2001).

14 Paigen, K. A miracle enough: the power of mice. Nature medicine 1, 215 (1995).

15 De Jong, M. & Maina, T. Of mice and humans: are they the same?—Implications

in cancer translational research. Journal of Nuclear Medicine 51, 501-504 (2010).

16 Perlman, R. L. Mouse models of human disease: An evolutionary perspective.

Evol Med Public Health 2016, 170-176, doi:10.1093/emph/eow014 (2016).

17 Sabounchi, N. S., Rahmandad, H. & Ammerman, A. Best-fitting prediction

equations for basal metabolic rate: informing obesity interventions in diverse

populations. Int J Obes (Lond) 37, 1364-1370, doi:10.1038/ijo.2012.218 (2013).

18 Blagosklonny, M. V. Big mice die young but large animals live longer. Aging

(Albany NY) 5, 227-233, doi:10.18632/aging.100551 (2013).

Page 239: Determining the Oncological and Immunological Effects of

202

19 Balmain, A. & C. Harris, C. Carcinogenesis in mouse and human cells: parallels

and paradoxes. Carcinogenesis 21, 371-377 (2000).

20 Greenberg, R. A., Allsopp, R. C., Chin, L., Morin, G. B. & DePinho, R. A.

Expression of mouse telomerase reverse transcriptase during development,

differentiation and proliferation. Oncogene 16, 1723 (1998).

21 Smith, V., Wirth, G. J., Fiebig, H. H. & Burger, A. M. Tissue microarrays of

human tumor xenografts: characterization of proteins involved in migration and

angiogenesis for applications in the development of targeted anticancer agents.

Cancer genomics & proteomics 5, 263-273 (2008).

22 Garrido-Laguna, I. et al. Tumor engraftment in nude mice and enrichment in

stroma- related gene pathways predict poor survival and resistance to

gemcitabine in patients with pancreatic cancer. Clinical cancer research : an

official journal of the American Association for Cancer Research 17, 5793-5800,

doi:10.1158/1078-0432.Ccr-11-0341 (2011).

23 Tratar, U. L., Horvat, S. & Cemazar, M. Transgenic mouse models in cancer

research. Frontiers in oncology 8 (2018).

24 Brock, R. M. et al. Patient derived xenografts expand human primary pancreatic

tumor tissue availability for ex vivo irreversible electroporation testing. Frontiers

in Oncology 10 (2020).

25 Sieren, J. C. et al. Development and translational imaging of a TP53 porcine

tumorigenesis model. The Journal of clinical investigation 124, 4052-4066

(2014).

Page 240: Determining the Oncological and Immunological Effects of

203

26 Ganderup, N. C., Harvey, W., Mortensen, J. T. & Harrouk, W. The minipig as

nonrodent species in toxicology—where are we now? International journal of

toxicology 31, 507-528 (2012).

27 Swindle, M. & Smith, A. Swine in the Laboratory: Surgery. Anesthesia, Imaging,

and Experimental Techniques, 40-42 (2007).

28 Swindle, M., Makin, A., Herron, A., Clubb Jr, F. & Frazier, K. Swine as models in

biomedical research and toxicology testing. Veterinary pathology 49, 344-356

(2012).

29 Segatto, N. V. et al. The oncopig cancer model as a complementary tool for

phenotypic drug discovery. Frontiers in pharmacology 8, 894 (2017).

30 Maher, B. Tissue engineering: How to build a heart. Nature News 499, 20 (2013).

31 Crile, G. W., Hosmer, H. R. & Rowland, A. F. The electrical conductivity of animal

tissues under normal and pathological conditions. American Journal of

Physiology-Legacy Content 60, 59-106 (1922).

32 Davalos, R. V., Mir, L. M. & Rubinsky, B. Tissue ablation with irreversible

electroporation. Ann Biomed Eng 33, 223-231 (2005).

33 Maor, E., Ivorra, A., Leor, J. & Rubinsky, B. The effect of irreversible

electroporation on blood vessels. Technol Cancer Res Treat 6, 307-312 (2007).

34 Garcia, P. A. et al. Non-thermal irreversible electroporation (N-TIRE) and

adjuvant fractionated radiotherapeutic multimodal therapy for intracranial

malignant glioma in a canine patient. Technol Cancer Res Treat 10, 73-83

(2011).

Page 241: Determining the Oncological and Immunological Effects of

204

35 Neal, R. E., 2nd et al. Successful treatment of a large soft tissue sarcoma with

irreversible electroporation. Journal of clinical oncology : official journal of the

American Society of Clinical Oncology 29, e372-377,

doi:10.1200/JCO.2010.33.0902 (2011).

36 Li, W., Fan, Q., Ji, Z., Qiu, X. & Li, Z. The Effects of Irreversible Electroporation

(IRE) on Nerves. PLOS ONE 6, e18831, doi:10.1371/journal.pone.0018831

(2011).

37 Lee, K. et al. Engraftment of human iPS cells and allogeneic porcine cells into

pigs with inactivated RAG2 and accompanying severe combined

immunodeficiency. Proc Natl Acad Sci U S A 111, 7260-7265,

doi:10.1073/pnas.1406376111 (2014).

38 Choi, Y. J. et al. Recombination activating gene-2(null) severe combined

immunodeficient pigs and mice engraft human induced pluripotent stem cells

differently. Oncotarget 8, 69398-69407, doi:10.18632/oncotarget.20626 (2017).

39 Choi, Y. J. et al. Partial loss of interleukin 2 receptor gamma function in pigs

provides mechanistic insights for the study of human immunodeficiency

syndrome. Oncotarget 7, 50914-50926, doi:10.18632/oncotarget.10812 (2016).

40 Kang, J. T. et al. Biallelic modification of IL2RG leads to severe combined

immunodeficiency in pigs. Reprod Biol Endocrinol 14, 74, doi:10.1186/s12958-

016-0206-5 (2016).

41 Lei, S. et al. Increased and prolonged human norovirus infection in RAG2/IL2RG

deficient gnotobiotic pigs with severe combined immunodeficiency. Sci Rep 6,

25222, doi:10.1038/srep25222 (2016).

Page 242: Determining the Oncological and Immunological Effects of

205

42 Lei, S. et al. Increased and prolonged human norovirus infection in RAG2/IL2RG

deficient gnotobiotic pigs with severe combined immunodeficiency. Scientific

reports 6, 25222 (2016).

43 Lee, K. et al. Engraftment of human iPS cells and allogeneic porcine cells into

pigs with inactivated RAG2 and accompanying severe combined

immunodeficiency. Proceedings of the National Academy of Sciences 111, 7260-

7265 (2014).

44 Basel, M. T. et al. Human xenografts are not rejected in a naturally occurring

immunodeficient porcine line: a human tumor model in pigs. BioResearch open

access 1, 63-68 (2012).

45 Jain, R., Fischer, S., Serra, S. & Chetty, R. The use of cytokeratin 19 (CK19)

immunohistochemistry in lesions of the pancreas, gastrointestinal tract, and liver.

Applied Immunohistochemistry & Molecular Morphology 18, 9-15 (2010).

46 Halter, R. J., Schned, A., Heaney, J., Hartov, A. & Paulsen, K. D. Electrical

properties of prostatic tissues: I. Single frequency admittivity properties. the

Journal of Urology 182, 1600-1607 (2009).

47 Guo, S. et al. Enhanced Electric Pulse Technology for the Ablation of Pancreatic

Cancer. Advances in Pancreatic Cancer, 115 (2018).

48 Moschidis, A. et al. Gemcitabine in combination with Oxaliplatin in Pancreatic

(PAN-02) tumor bearing mice. Journal of Clinical Oncology 22, 4153-4153

(2004).

49 Hofmeister, V., Schrama, D. & Becker, J. C. Anti-cancer therapies targeting the

tumor stroma. Cancer Immunology, Immunotherapy 57, 1-17 (2008).

Page 243: Determining the Oncological and Immunological Effects of

206

50 Ma, W. et al. Tumor-stroma ratio is an independent predictor for survival in

esophageal squamous cell carcinoma. Journal of Thoracic Oncology 7, 1457-

1461 (2012).

51 Dittmer, J. & Leyh, B. in Seminars in cancer biology. 3-15 (Elsevier).

52 Torres, M. P. et al. Novel pancreatic cancer cell lines derived from genetically

engineered mouse models of spontaneous pancreatic adenocarcinoma:

applications in diagnosis and therapy. PloS one 8, e80580 (2013).

53 Zhou, R., Xu, X., Liu, M., Wu, X. & Li, R. Immunophenotypes of ductal epithelial

cells in advanced pancreatic ductal adenocarcinoma. Digestion 99, 247-251

(2019).

54 Ivey, J. W. et al. Targeted cellular ablation based on the morphology of malignant

cells. Scientific reports 5, 17157 (2015).

55 Maor, E., Ivorra, A. & Rubinsky, B. Non thermal irreversible electroporation: novel

technology for vascular smooth muscle cells ablation. PloS one 4, e4757 (2009).

56 Murovec, T., Sweeney, D. C., Latouche, E., Davalos, R. V. & Brosseau, C.

Modeling of transmembrane potential in realistic multicellular structures before

electroporation. Biophysical journal 111, 2286-2295 (2016).

57 Lee, K. et al. Piglets produced from cloned blastocysts cultured in vitro with

GM‐CSF. Molecular reproduction and development 80, 145-154 (2013).

58 Yuan, L., Jobst, P. M. & Weiss, M. in Gnotobiotics 349-368 (Elsevier, 2017).

59 Abcam. (Abcam Corporation Cambridge, 2005).

Page 244: Determining the Oncological and Immunological Effects of

207

60 Bhonsle, S. et al. Characterization of irreversible electroporation ablation with a

validated perfused organ model. Journal of Vascular and Interventional

Radiology 27, 1913-1922. e1912 (2016).

61 Bhonsle, S., Lorenzo, M. F., Safaai-Jazi, A. & Davalos, R. V. Characterization of

Nonlinearity and Dispersion in Tissue Impedance During High-Frequency

Electroporation. IEEE Transactions on Biomedical Engineering 65, 2190-2201

(2017).

62 Beitel-White, N., Bhonsle, S., Martin, R. & Davalos, R. V. in 2018 40th Annual

International Conference of the IEEE Engineering in Medicine and Biology

Society (EMBC). 4170-4173 (IEEE).

63 Geddes, L. A. & Baker, L. E. The specific resistance of biological material—a

compendium of data for the biomedical engineer and physiologist. Medical and

biological engineering 5, 271-293 (1967).

64 Clark, D., Greenwell, J., Harper, A., Sankey, A. M. & Scratcherd, T. The electrical

properties of resting and secreting pancreas. The Journal of physiology 189, 247-

260 (1967).

65 Duck, F. A. Physical properties of tissues: a comprehensive reference book.

(Academic press, 2013).

66 McCann, H., Pisano, G. & Beltrachini, L. Variation in reported human head tissue

electrical conductivity values. Brain topography 32, 825-858 (2019).

67 Stoy, R. D., Foster, K. R. & Schwan, H. P. Dielectric properties of mammalian

tissues from 0.1 to 100 MHz; a summary of recent data. Physics in Medicine &

Biology 27, 501 (1982).

Page 245: Determining the Oncological and Immunological Effects of

208

68 Neal, R. E. et al. In vivo characterization and numerical simulation of prostate

properties for non‐thermal irreversible electroporation ablation. The Prostate 74,

458-468 (2014).

Page 246: Determining the Oncological and Immunological Effects of

209

5.10 Supplemental Figure

Figure 5-9 Supplemental Figure 5.S1. Genotyping of piglets carrying modified IL2RG and RAG2 gene. Six piglets were born and analyzed. During PCR analysis, RAG2 was not amplified from the Piglet #2 DNA sample, suggesting a large deletion. No wild-type allele was identified.

Page 247: Determining the Oncological and Immunological Effects of

210

Chapter 6. Feasibility of Histotripsy Pancreas Ablation in a Large Pig Model

Alissa Hendricks-Wenger, Jessica Gannon, Neha Singh, Lauren Arnold, Michael Edwards, Khan Mohammad Imran, Margaret Nagai-Singer, Benjamin Tintera, Hannah Sheppard, Joan Vidal-Jove, David Luyimbazi, Mishal Mendiratto-Lala, Martha Larson,

Sheryl Coutermarsh-Ott, Irving C Allen, Eli Vlaisavljevich

This chapter is excerpted from a manuscript in preparation for submission to Ultrasound in Medicine and Biology.

Page 248: Determining the Oncological and Immunological Effects of

211

6.1 Abstracts

Pancreatic cancer is one of the most lethal forms of cancer, with a 5 year-survival rate

of 9%. To address this, many new therapeutic modalities are being developed. One

modality is the focused-ultrasound therapy Histotripsy, a non-invasive, non-ionizing, and

non-thermal ablation modality that has been shown to be effective in treating hepatic

malignancies. However, when translating an ultrasonic modality to the pancreas there

are limitations that need to be considered, including 1) achieving an acoustic window

and 2) generating an ablation lesion without causing moderate or severe complications.

For this study, we used healthy pigs in order to test the efficacy of our custom 32

element, 500kHz histotripsy transducer that is comparable to human scale devices. To

obtain an ideal acoustic window, a pilot study was conducted using pigs that had been

fasted for at least 12 hours. While we were able to use our system to generate a bubble

cloud and lesion within the liver, we were unable to get an acoustic window for targeting

the pancreas. Using a laxative and simethicone custard in combination with fasting, the

gas within the porcine gastrointestinal system was reduced. This allowed for easier

ultrasound targeting of the pancreas, generation of a bubble cloud within the pancreas,

and creation of a clear lesion. Acute necropsy demonstrated mild bruising to intestines

and the stomach that was near the focus as well as in some loops of pre-focal bowel.

Following a group (n=4) for 1-week post treatment through behavioral observations,

diet, and blood work, we did not find signs of any complications. At 1 week on contrast

enhanced CT imaging and necropsy, there were no signs of bruising or abnormalities

Page 249: Determining the Oncological and Immunological Effects of

212

on any organs. Overall, these results suggest that our histotripsy system and degassing

protocol creates the opportunity to target the porcine pancreas in future studies.

6.2 Keywords:

Pancreas, Pancreatic Cancer, Histotripsy, Porcine, Ultrasound Imaging

Page 250: Determining the Oncological and Immunological Effects of

213

6.3 Introduction

Pancreatic cancer is considered a fatal malignancy with a 5-year survival rate lower

than 9% and the majority of patients dying within 6 months of diagnosis [1]. Most

complications that contribute to this disease are associated with a late diagnosis, rapid

local progression with vascular compromise, and metastasis. The current gold standard

for treatment includes surgery [2]. However, the location of the tumor is a strong

prognostic factor, and its proximity to vital vascular structures and local infiltration into

the adjacent mesenteric fat are major limitations precluding resection in a large number

of patients [3]. Despite improved outcomes with current frontline therapeutics, a high

incidence of refractory disease from chemo- and radio-resistance, as well as local and

distant recurrence, still pose significant challenges [2]. There are a multitude of tumor

ablation methods including radiofrequency (RFA), microwave (MWA), cryoablation, high

intensity focused ultrasound (HIFU), and irreversible electroporation (IRE) [4-6].

However, only HIFU and IRE have been developed as stand-alone or combination

therapeutic options for pancreatic cancer, given the high complication risk and technical

challenges of RFA and MWA [7, 8].

IRE and HIFU have shown some improvement over standard of care in terms of

morbidity, tissue conservation, improved surgical accessibility, and decreased

hospitalization [9, 10]. However, these strategies can be associated with significant

complications, and many patients are not candidates due to local tumor progression,

tumor size, and location near critical structures [10, 11]. HIFU, among other thermal

Page 251: Determining the Oncological and Immunological Effects of

214

ablation methods, runs a high risk for thermal injury to critical structures near the

ablation zone, particularly pancreatic duct injury. This damage can result in a leak and

severe pancreatitis or even stenosis with resultant obstructive pancreatitis [9].

Additionally, the lack of real-time treatment feedback limits the ability to create precise

ablation zones. The invasive needle puncture used in IRE, inability to treat large (>3 cm

diameter) or multiple tumor nodules, and incomplete ablation near major vessels are

additional limitations. All of these limitations render these treatment modalities as

challenging for clinical use [9, 10]. As such, there remains an unmet need for a new

ablation method that can address the limitations of currently available ablation methods

for pancreatic cancer.

Histotripsy is a non-invasive, non-ionizing, and non-thermal focused ultrasound

ablation method that destroys tissue through the precise control of acoustic cavitation

[12, 13]. Unlike thermal HIFU, histotripsy disrupts tissue through mechanical effects

(i.e., cavitation) [13]. Using high-pressure pulses applied by an external transducer, a

cavitation “bubble cloud” is generated at the target [13, 14], and the expansion and

collapse of the bubbles ablates the tissue into an acellular homogenate [13, 14]. Since

histotripsy is non-thermal, it does not have the many limitations associated with thermal

ablation [15, 16]. Histotripsy can produce consistent and rapid ablation, even in close

proximity to major vessels, with millimeter precision and well-defined boundaries

(<2mm) [17]. The high accuracy of histotripsy is a direct result of the non-thermal

mechanism, which results in a “binary” treatment outcome with sharp boundaries and

millimeter precision. This is a significant improvement upon thermal ablation methods

that do not have well-defined margins and are often surrounded by a centimeter or more

Page 252: Determining the Oncological and Immunological Effects of

215

of partially treated tissue due to thermal spread [17, 18]. Furthermore, histotripsy can

preserve critical structures such as large vessels and bile ducts due to the higher

mechanical strength of these structures [17, 18]. Additional benefits include real-time

imaging feedback [19] and the ability to cover tumor nodules of arbitrary size and shape

[20]. Due to these features, histotripsy has the potential to overcome the weaknesses of

current treatment methods and benefit many diverse diseases [21].

Previous in vivo animal studies have investigated histotripsy’s use for precise and

complete liver ablation. These studies also showed that histotripsy can generate tissue

selective ablation in which the target tissue is completely destroyed into an acellular

homogenate, while critical structures (vessels and bile ducts) are preserved [17]. These

initial studies led to the development of a clinical prototype histotripsy system, which

was subsequently tested in a pig model, to show the ability of producing clinically

relevant ablation zones in livers that are closer to human size. Additionally, these

preclinical results led to a Phase I clinical trial in the FUS Ablation Oncology Unit of

Hospital University Mutua Terrassa (Barcelona, Spain), led by Dr. Joan Vidal Jove, one

of our key clinical collaborators. Favorable results from the trial demonstrated that

histotripsy can non-invasively produce well-tolerated treatments and rapid involution of

ablation zones without significant device-related adverse events [22].

Previous work has shown the feasibility of focused ultrasound (FUS) for targeting the

pancreas. For example, a series of clinical studies have been performed on non-

resectable pancreatic tumors (Stages III/IV) in the FUS Ablation Oncology Unit of

Hospital University Mutua Terrassa in Barcelona, Spain [22]. Results showed a

sufficient acoustic window available to apply HIFU to pancreatic tumors under

Page 253: Determining the Oncological and Immunological Effects of

216

ultrasound guidance. Clinical responses (thermal ablation achieved) were 82% in all

cases, sustained at 8 weeks of the procedure, with 12 complete responses (21% of

patients, 10 stage III, 2 stage IV) at the end of a combined HIFU and chemotherapy

regime. However, results also showed major complications due to the thermal

mechanism that included pancreatitis with GI bleeding and skin burning grade III that

required plastic surgery. Overall, these results showed that FUS can target the

pancreas non-invasively under ultrasound guidance with the potential to achieve

effective treatments outcomes. Results also showed limitations of HIFU due to the

thermal mechanism, including injury to critical structures and overlying tissue. We

expect histotripsy to overcome these limitations due to its non-thermal mechanism,

allowing for safer, more precise, and more effective pancreatic ablation.

While prior studies highlight the potential of histotripsy for non-invasive tumor

ablation in other organs, significant work remains to establish histotripsy’s potential in

pancreatic tumor ablation. The pancreas poses unique challenges for ablation

modalities due to the tissue characteristics and risk of damage to nearby critical

structures. In this study, we investigate the feasibility of histotripsy in the treatment of

healthy pancreatic tissue in an in vivo pig model.

6.4 Materials and Methods

6.4.1 Animal Monitoring and Anesthesia

Page 254: Determining the Oncological and Immunological Effects of

217

Male pigs (6-8 weeks old, 5-8kg; Virginia Tech Swine Farm, Blacksburg, VA)

were monitored three times during their first week to acclimate to their new housing, and

then daily after treatment in accordance with Virginia Tech IACUC protocols. At each

check, general health was monitored looking for changes in behavior, diet, and skin

lesions. Blood was collected into green and lavender vacutainer tubes at baseline and

post-treatment days 1 and 5. Tubes were submitted to the Virginia Maryland Animal

Laboratory Services (Blacksburg, VA) for analysis for complete blood cell and

pancreatic enzyme panels. General workflow shown in Figure 1.

For treatment, animals were sedated by using an intramuscular injection of the

cocktail of Telazol, Ketamine, and Xylazine administered in intervals approximately 15-

20 minutes apart, or when the animal began to demonstrate increased muscle tone.

Animals’ heart rate, blood oxygen levels, respiratory rate, and temperature were

monitored during treatment by trained personnel. Until fully recovered, animals were

visually monitored for 2-3 hours in recovery kennels before being returned to their

standard housing.

Figure 6-1Study timeline demonstrating the relative time for treatment, custard feeding, blood draws, and CT imaging.

Page 255: Determining the Oncological and Immunological Effects of

218

6.4.2 Histotripsy Systems and Pressure Calibrations

Histotripsy was performed in vivo using a custom 32-element, 500 kHz large

animal histotripsy transducer with a geometric focus of 78 mm, an elevational aperture

size of 112 mm, and a transverse aperture size of 128 mm. Corresponding f-numbers

include 0.61 (transverse) and 0.70 (elevational). To generate short therapy pulses <2

cycles, the transducer was controlled via a custom high-voltage pulser with a field-

Figure 6-2 HistoSonics cart with 500 kHz transducer and coaxial US imaging probe mounted onto a micropositioner (A). Freehand imaging by radiologist to identify acoustic window (B). UMC bowl with therapy transducer submerged via adjustable arm (C).

Page 256: Determining the Oncological and Immunological Effects of

219

programmable gate array (FPGA) board (Altera DE0-Nano Terasic Technology, Dover,

DE, USA) programmed for histotripsy therapy pulsing. The transducer was mounted

onto a prototype clinical histotripsy treatment cart (HistoSonics, Ann Arbor, MI, USA)

consisting of an ultrasound imaging system, robotic micro-positioner, and customized

software to create volumetric ablations with histotripsy. A central hole through the

therapy transducer allowed a 3 MHz curvilinear imaging probe (Model C5-2, Analogic

Corp., Peabody, MA) to be coaxially aligned to enable real-time treatment guidance and

monitoring. [24, 25]. During histotripsy treatments, the transducer was triggered from

the HistoSonics software while being powered by a high voltage DC power supply

(GENH750W, TDK-Lambda), and the system was controlled using a custom user-

interface operated through MATLAB (MathWorks). The overall setup for the treatment is

illustrated in Figure 2A.

6.4.3 Hydrophone Focal Pressure Measurements and Beam Profiles

Focal pressure waveforms for the 500 kHz transducer were collected using a high

sensitivity rod hydrophone (HNR-0500, Onda Corporation, Sunnyvale, CA, USA) and a

custom-built fiber optic probe hydrophone (FOPH) [23, 24] in degassed water at the

focus of the transducer. 1D focal beam profiles of the transducer were measured in the

lateral, elevational, and axial directions with the rod hydrophone at a peak negative

pressure (p-) of ~1.8 MPa. The FOPH directly measured focal pressures up to a p- limit

of ~20 MPa due to cavitation on the tip of the FOPH fiber. Pressure waveforms at higher

p- were then estimated by a linear extrapolation [25]. All waveforms were captured at a

Page 257: Determining the Oncological and Immunological Effects of

220

sample rate of 500MS/s using a Tektronix TBS2000 series oscilloscope with the

waveforms averaged over 128 pulses and recorded in MATLAB. This procedure with is

described in previous studies further details [25, 26].

6.4.4 Histotripsy In Vivo Pancreas Ablation Procedure

The feasibility of histotripsy for pancreas ablation was tested in vivo using

healthy, young pig. This custom system, which has been used in previous large animal

in vivo histotripsy studies [27-29], consists of the 500 kHz histotripsy transducer with co-

axially aligned ultrasound imaging connected to an articulating arm with 4 degrees of

freedom (Company Source of Arm). Six male pigs were purchased at 6 weeks of age

from the pig farm at Virginia Tech. Once received, animals were monitored three times

during their first week to acclimate to their new holding pins, and then daily after

treatment, in accordance with Virginia Tech IACUC protocols. At each check, general

health was monitored, and blood was collected into vacutainer tubes at baseline and

post-treatment day 5.

For the 4 days leading to treatments, the animals were fed a sweetened custard

once daily with simethicone (2 mL/pig) and bisacodyl (0.3 mg/kg) in order to minimize

the intestinal contents and gas present on the day of treatment. Pigs were fasted from

food after their evening meal the day before treatment, and were given an additional

custard 2 hours before the first treatment of the morning. Immediately before treatment,

hair was removed over the abdomen by shaving with electric razor and then Nair

(Naircare, Ewing, NJ, USA) was applied for 10 minutes before being removed by wiping

Page 258: Determining the Oncological and Immunological Effects of

221

off with a wet towel. For treatment, pigs were anesthetized with an intramuscular

injection of the cocktail TKX (Telazol-Ketamine-Xylazine) and placed in the dorsal

recumbent position on the treatment table with feet loosely restrained to maintain a

stable position (Fig.2B). Heart rate, blood oxygen levels, and temperature were

monitored for all pigs during treatment by trained personnel.

Before treatment, freehand ultrasound imaging was performed by a board-

certified veterinary radiology (M.L.) or veterinary radiology resident (M.E.) to identify the

pancreas and determine the desired treatment location using both a linear ultrasound

imaging probe with a frequency range of 10-18 MHz (L18-10L30H-4, Telemed,

Lithuania, EU) and a 3 MHz curvilinear imaging probe (Model C5-2, Analogic Corp.,

Peabody, MA) (Fig.2B). Custom Ioban (3M) drapes with a hole large enough to

establish an acoustic window were attached to the pig’s abdomen. A plastic ultrasound

mediated coupling (UMC) bowl, with the base removed attached to an articulating arm,

was mounted to the surgical table. The bowl was then manually adjusted with the

articulating arm to align the hole of the bowl with the acoustic treatment window. Excess

drape was carefully pulled into the bowl and clamped to the upper rim. Before

treatment, a water tub was filled with water and degassed for ~2 hours, with this water

being used to fill the UMC bowl for ultrasound coupling. The therapy transducer and

coaxial imaging probe were mounted onto a mechanical arm containing a

micropositioner (Histosonics Inc., Ann Arbor, MI) [28], was manually positioned to

aligned with the acoustic window identified during freehand imaging (Fig.2C). A bubble

cloud was generated in degassed water prior to treatment to align the focus of the

transducer with system’s imaging screen

Page 259: Determining the Oncological and Immunological Effects of

222

For treatment, the coaxial imaging probe was used to align the focus of the

transducer to the targeted location. The transducer was then turned on and histotripsy

was applied to the target using single cycle pulses and a pulse repetition frequency

(PRF) of 500Hz. The cavitation threshold within the tissue was determined by slowly

increasing the pressure until a robust bubble cloud was generated. The transducer

swept through a 1 – 1.5 cm diameter spherical ablation volume delivering 500 pulses

per treatment point with a peak negative pressure of ~21 MPa. Throughout the entire

procedure, ultrasound imaging was used to monitor the treatment in real-time. To

ensure a complete ablation of the targeted pancreas, a second treatment to the exact

same volume was immediately done after the first treatment. After treatment, the

therapy transducer was removed and freehand ultrasound imaging was performed

assess for any tissue damage. Animals in the acute group (n=2) were immediately

euthanized by an intracardiac injection of pentobarbital sodium and phenytoin sodium

(Euthasol) according to protocol. Animals in the 1-week group were anesthetized and

euthanized after CT. A full necropsy was conducted post-treatment by a board-certified

veterinary pathologist (S.C.O.).

6.4.5 Computed Tomography Imaging

Anesthetized pigs were placed in a supine position in a veterinary CT scanner

(Toshiba Aquilion 16-slice) for 1 week post treatment imaging. CTs were performed 1

week after treatment, based on prior studies that show that moderate and severe post-

Page 260: Determining the Oncological and Immunological Effects of

223

operative complications occur in this window [30]. Images were analyzed by a board-

certified veterinary radiologist (M.L.) and veterinary radiology resident (M.E.).

6.4.6 Necropsy & Histological Analysis

The acute and 1-week-survival groups animals’ necropsy and tissue harvest was

preformed either immediately and 1-week post-treatment, respectively, by a board-

certified veterinary pathologist (S.C.O.). Gross damage to internal organs was

determined with inspection in situ and ex vivo with bread slices spaced approximately

1cm apart. Bread slicing of pancreatic tissue was performed after fixation due to the

friable nature of the pancreatic tissue. Pancreatic tissue and any tissue with noted gross

changes were visually inspected and fixed in 10% formalin for at least 24 hours before

sectioning, embedding, and staining. All tissues were stained using hematoxylin and

eosin (H&E) to assess for histotripsy damage to the tissue parenchyma and any other

changes in structure and density of collagen and other tissue structures within the

ablation volume. Analysis was led by a board-certified veterinary pathologist (S.C.O.) to

ensure accuracy and avoid biases.

6.5 Results

6.5.1 Establishing Safe, Non-Invasive Therapeutic Protocol

Targeting the liver with our histotripsy system clearly showed a describable bubble

cloud (Fig.3A) and on gross necropsy showed a clear ablation region (Fig.3B).

Page 261: Determining the Oncological and Immunological Effects of

224

However, there was a more difficult time getting clear ultrasound images of the bubble

cloud within the pancreas. In the pilot group, in which the pigs were only fasted for ~12

hours before treatment, there was difficulty imaging the pancreas with the off-set

imaging (Fig.3C). When the pigs were fed the laxative, simethicone custard on the

prescribed regime, the dynamic nature of the bubble cloud was visible on ultrasound.

Due to the hyperechoic nature of the pancreas relative to the bubble cloud, it is difficult

to discern the bubble cloud in still frame images of the pancreas (Fig.3D).

Figure 6-3 Bubble cloud formed in liver on US imaging from HistoSonics system (pig 1, 8-6)(A). No bubble cloud visualized on US imaging and did not clearly identify the pancreas without pudding (pig 6, 8-13)(B). Bubble cloud generated inside pancreas where pig was fed pudding prior to histotripsy treatment (28-53) (C).

Page 262: Determining the Oncological and Immunological Effects of

225

Intestinal gas interference during treatment prevented ideal imaging quality and

bubble formation (Fig.3C) leading to pre-focal intestinal bruising (Fig.4A-B). Bruising

was more highly concentrated in the more gaseous large intestine, indicated by the

green arrow, and near the targeted region, indicated by the blue arrows. Post treatment

H&E histopathology indicated that from a sample of the bruised colon, the muscularis

Figure 6-4 Intestinal gas interference with treatment prevented ideal imaging quality and bubble formation during treatment leading to pre-focal intestinal bruising (A-B) more highly concentrated in the more gaseous large intestine, indicated by the green arrow, and near the targeted region, indicated by the blue arrows. Post treatment H&E histopathology indicated that from a sample of the bruised colon, the muscularis was not damaged but there was a large amount of ablation within the submucosa (C-D).

Page 263: Determining the Oncological and Immunological Effects of

226

was not damaged but there was a large amount of ablation within the submucosa

(Fig.4C-D).

When using the custard as part of the bowel preparation, the improved imaging

(Fig.5A) allowed for improved ablation. There was evident lesion generation within the

pancreas within the laxative, simethicone custard group of pigs. Post-treatment US

imaging (Fig.5B) clearly shows a hypoechoic region, consistent with a histotripsy

ablation, in the same target location. In these animals, while we did have off target

effects such as bruising to the intestinal tissues (Fig.6), this was reduced from the pilot

group that was not fed the custard mix.

6.5.2 Physiological Outcomes

All animals included in the 1 week-survival group (n=4) recovered well from

treatment, and were returned to standard housing with their normal, unrestricted diet

within 4 hours of treatment. Over the week following treatment, the animals had no

signs of debilitating side effects. Specifically, there was no note of: fatigue, pain,

vomiting, diarrhea, or discolored stool. All treated animals maintained normal weight,

Figure 6-5 US imaging of pancreas before histotripsy (A) and after treatment with clear hypoechoic region identifying the targeted ablated volume (B). Dotted red circle indicated the approximate region treated

Page 264: Determining the Oncological and Immunological Effects of

227

eating and drinking habits, levels of activity, and socialization. WBC, amylase, and

lipase levels of all animals were unaffected by histotripsy treatment 1 and 5 days post-

treatment compared to their baseline values determined 5 days prior to treatment

(Fig.7). No abnormalities were noted on the complete blood cell panel. From the 1-week

post treatment group, CT showed that there was no free fluid in the abdomen (Fig.8).

Chronic necropsy demonstrated that there was no gross damage to any organ in situ

and there was no free fluid or intestinal contents in the abdomen (Fig.9). There was no

splenomegaly, and bread slices to liver and spleen tissues further confirmed no off-

target ablation within those organs. Bread slices of formalin fixed pancreatic tissues did

not reveal any gross lesions. Histology from the 1-week necropsy showed no notable

lesions, hemorrhage, or inflammation within the pancreas (Fig.10).

Figure 6-6 With laxative, simethicone, and custard pretreatment protocol, there was signifcatly less bruising in preforcal bowel (A) indicated by the green arrow. Bruising around the treatment area was more concentrated than on the bowel, but was still less intense than the acute group (B), bruising indicated by blue arrow.

Page 265: Determining the Oncological and Immunological Effects of

228

0 5 10

0

10

20

30

40

50

White Blood Cell

Days Post Arrival

WB

Cs

(x

10^

3 c

ell

s/u

L) Treatment

0 5 10

0

500

1000

1500

2000

Amylase

Days Post Arrival

Am

yla

se (

U/L

)

Treatment

0 5 10

0

5

10

15

20

Lipase

Days Post Arrival

Lip

as

e (

U/L

)

Treatment Pig 1

Pig 2

Pig 3

Pig 4

Figure 6-8 White blood cell, amylase, and lipase serum levels for 1-week survival animals.

Figure 6-7 CT taken in the dorsal recumbent position (A) one week post treatment shows no free fluid in the peritoneal cavity (B)

Page 266: Determining the Oncological and Immunological Effects of

229

Figure 6-10 Necropsy reviled no gross damage within the abdomen in situ (A-B). When excised in tota no damage was found on the exterior of the liver (C-D), the spleen (E), nor the pancreas (F-G).

Figure 6-9 1-week post treatment there was no signs of morbidity within the pancreatic tissue. Images representative of two animals.

Page 267: Determining the Oncological and Immunological Effects of

230

6.6 Discussions

In this study, we investigated the feasibility of using histotripsy for pancreatic tissue

ablation in an in vivo porcine model. Targeting intraabdominal organs, such as the

pancreas, with ultrasonic imaging and therapies requires additional planning and

investigation to determine the ideal preoperative and operative procedures that can

minimize the effects of pre-focal, acoustically opaque gas pockets within the

gastrointestinal tract. Given that the two notable benefits of histotripsy as an ablation

modality are that it is non-invasive as well as ultrasound imaging guided, the focus of

this study was to determine the feasibility of this approach for targeting the pancreas. A

prior porcine study utilizing HIFU to treat the pancreas was able to perform the

procedure without the use of nasogastric tube or other more invasive technique to

remove the gas from the gastrointestinal tract [31]. However, given HIFU’s thermal

mechanism of ablation, they were limited to using MR imaging for planning due to the

lack of ultrasonic methods for measuring ablative temperature change. In this study, a

combination of simethicone, laxative, and custard, previously reported non-invasive

methods for degassing the stomach and intestines [31], allowed for histotripsy to be

applied solely with ultrasound guidance (Fig.5).

Potential off-target effects are a significant hurdle for therapeutic ablation techniques

like histotripsy, particularly for structures like the pancreas which are surrounded by

critical structures. Results from this study suggest that histotripsy can be used to

effectively target the pancreas without causing damage to nearby, critical structures.

While off-target intestinal bruising was identified (Fig.6), there were no signs of

Page 268: Determining the Oncological and Immunological Effects of

231

pancreatitis (Fig.7), intestinal perforation, intraabdominal bleeding, or pancreatic fistulas

(Fig.8 and Fig.9). Determining if an ablation therapy causes pancreatitis is important

due to the need to treat margins without causing negative side effects. Given that the

porcine pancreas is similar in anatomy and presentation of acute pancreatitis to humans

[32-34], they make an appropriate model for this style of investigation. A previous

porcine study testing the use of IRE within the pancreas showed a rise in WBCs,

amylase, and lipase as well as anorexia after treatment [35]. While this pilot study for

using histotripsy to target the pancreas is preliminary for targeting the pancreas, the

safety profile is evident. By being non-invasive, we did not risk infection. By propagating

the therapeutic effect directly to the targeted region, we did not agitate additional

pancreatic stroma to the point of causing acute pancreatitis.

In addition to the concern of pancreatitis previously reported in association with other

ablation modalities, there is an additional concern that histotripsy could destabilize the

mechanical structure of the pancreas resulting in a pancreatic fistula. The 2016

International Study Group of Pancreatic Fistula redefined a pancreatic fistula as post-

operative drainage from the pancreas that results in a sequelae that requires clinical

intervention to prevent mortality [36, 37]. From the 4 pigs that were treated with

histotripsy in the study and survived for 1 week to determine post-treatment side effects,

there were no changes to behavior or diet and no signs on CT (Fig.8) or necropsy

(Fig.9) to indicate that a pancreatic fistula was formed.

Page 269: Determining the Oncological and Immunological Effects of

232

6.7 Conclusion

This study demonstrates the feasibility of histotripsy for non-invasive ablation within

the pancreas. Overall, this study demonstrated that histotripsy can be used to treat

targets within the pancreas without causing moderate nor severe side effects. Future

work will aim the further optimize histotripsy for pancreas ablation and determine the

potential of histotripsy to ablate pancreatic tumors.

6.8 Acknowledgements

Thanks for Kristen Eden, DVM, PhD for providing additional necropsy support for 2

animals when S.C.O. was unavailable.

Page 270: Determining the Oncological and Immunological Effects of

233

6.9 References

[1] R. L. Siegel, K. D. Miller, and A. Jemal, "Cancer statistics, 2020," CA: a cancer

journal for clinicians, vol. 70, no. 1, pp. 7-30, 2020.

[2] E. G. Chiorean and A. L. Coveler, "Pancreatic cancer: optimizing treatment

options, new, and emerging targeted therapies," Drug design, development and

therapy, vol. 9, pp. 3529-45, 2015, doi: 10.2147/DDDT.S60328.

[3] A. Artinyan, P. A. Soriano, C. Prendergast, T. Low, J. D. Ellenhorn, and J. Kim,

"The anatomic location of pancreatic cancer is a prognostic factor for survival,"

HPB : the official journal of the International Hepato Pancreato Biliary

Association, vol. 10, no. 5, pp. 371-6, 2008, doi: 10.1080/13651820802291233.

[4] S. Paiella et al., "Radiofrequency ablation for locally advanced pancreatic cancer:

SMAD4 analysis segregates a responsive subgroup of patients," Langenbecks

Arch Surg, vol. 403, no. 2, pp. 213-220, Mar 2018, doi: 10.1007/s00423-017-

1627-0.

[5] H. Y. Ge et al., "High-intensity focused ultrasound treatment of late-stage

pancreatic body carcinoma: optimal tumor depth for safe ablation," Ultrasound

Med Biol, vol. 40, no. 5, pp. 947-55, May 2014, doi:

10.1016/j.ultrasmedbio.2013.11.020.

[6] Y. Keisari, "Tumor abolition and antitumor immunostimulation by physico-

chemical tumor ablation," Front Biosci (Landmark Ed), vol. 22, pp. 310-47, 2017.

Page 271: Determining the Oncological and Immunological Effects of

234

[7] A. Sofuni et al., "Safety trial of high-intensity focused ultrasound therapy for

pancreatic cancer," World J Gastroenterol, vol. 20, no. 28, pp. 9570-7, Jul 28

2014, doi: 10.3748/wjg.v20.i28.9570.

[8] K. Sugimoto et al., "Irreversible Electroporation for Nonthermal Tumor Ablation in

Patients with Locally Advanced Pancreatic Cancer: Initial Clinical Experience in

Japan," Intern Med, vol. 57, no. 22, pp. 3225-3231, Nov 15 2018, doi:

10.2169/internalmedicine.0861-18.

[9] A. M. Ierardi et al., "Percutaneous ablation therapies of inoperable pancreatic

cancer: a systematic review," Ann Gastroenterol, vol. 28, no. 4, pp. 431-9, Oct-

Dec 2015. [Online]. Available: https://www.ncbi.nlm.nih.gov/pubmed/26424487.

[10] M. D'Onofrio et al., "Percutaneous ablation of pancreatic cancer," World J

Gastroenterol, vol. 22, no. 44, pp. 9661-9673, Nov 28 2016, doi:

10.3748/wjg.v22.i44.9661.

[11] M. Linecker, T. Pfammatter, P. Kambakamba, and M. L. DeOliveira, "Ablation

Strategies for Locally Advanced Pancreatic Cancer," Dig Surg, vol. 33, no. 4, pp.

351-9, 2016, doi: 10.1159/000445021.

[12] J. E. Parsons, C. A. Cain, G. D. Abrams, and J. B. Fowlkes, "Pulsed cavitational

ultrasound therapy for controlled tissue homogenization," Ultrasound Med Biol,

vol. 32, no. 1, pp. 115-29, Jan 2006, doi: 10.1016/j.ultrasmedbio.2005.09.005.

[13] E. Vlaisavljevich, A. Maxwell, L. Mancia, E. Johnsen, C. Cain, and Z. Xu,

"Visualizing the Histotripsy Process: Bubble Cloud-Cancer Cell Interactions in a

Tissue-Mimicking Environment," Ultrasound Med Biol, vol. 42, no. 10, pp. 2466-

77, Oct 2016, doi: 10.1016/j.ultrasmedbio.2016.05.018.

Page 272: Determining the Oncological and Immunological Effects of

235

[14] Z. Xu, M. Raghavan, T. L. Hall, M. A. Mycek, and J. B. Fowlkes, "Evolution of

bubble clouds induced by pulsed cavitational ultrasound therapy - histotripsy,"

IEEE Trans Ultrason Ferroelectr Freq Control, vol. 55, no. 5, pp. 1122-32, May

2008, doi: 10.1109/TUFFC.2008.764.

[15] E. Vlaisavljevich et al., "Non-Invasive Liver Ablation Using Histotripsy: Preclinical

Safety Study in an In Vivo Porcine Model," Ultrasound Med Biol, vol. 43, no. 6,

pp. 1237-1251, Jun 2017, doi: 10.1016/j.ultrasmedbio.2017.01.016.

[16] Y. Kim, E. Vlaisavljevich, G. E. Owens, S. P. Allen, C. A. Cain, and Z. Xu, "In vivo

transcostal histotripsy therapy without aberration correction," Phys Med Biol, vol.

59, no. 11, pp. 2553-68, Jun 7 2014, doi: 10.1088/0031-9155/59/11/2553.

[17] E. Vlaisavljevich, Y. Kim, G. Owens, W. Roberts, C. Cain, and Z. Xu, "Effects of

tissue mechanical properties on susceptibility to histotripsy-induced tissue

damage," Phys Med Biol, vol. 59, no. 2, pp. 253-70, Jan 20 2014, doi:

10.1088/0031-9155/59/2/253.

[18] A. M. Lake, Z. Xu, J. E. Wilkinson, C. A. Cain, and W. W. Roberts, "Renal

ablation by histotripsy--does it spare the collecting system?," J Urol, vol. 179, no.

3, pp. 1150-4, Mar 2008, doi: 10.1016/j.juro.2007.10.033.

[19] T. Y. Wang, T. L. Hall, Z. Xu, J. B. Fowlkes, and C. A. Cain, "Imaging feedback of

histotripsy treatments using ultrasound shear wave elastography," IEEE Trans

Ultrason Ferroelectr Freq Control, vol. 59, no. 6, pp. 1167-81, Jun 2012, doi:

http://dx.doi.org/10.1109/TUFFC.2012.2307

10.1109/tuffc.2012.2307.

Page 273: Determining the Oncological and Immunological Effects of

236

[20] J. E. Lundt, S. P. Allen, J. Shi, T. L. Hall, C. A. Cain, and Z. Xu, "Non-invasive,

Rapid Ablation of Tissue Volume Using Histotripsy," Ultrasound Med Biol, vol. 43,

no. 12, pp. 2834-2847, Dec 2017, doi: 10.1016/j.ultrasmedbio.2017.08.006.

[21] E. Vlaisavljevich et al., "Image-guided non-invasive ultrasound liver ablation

using histotripsy: feasibility study in an in vivo porcine model," Ultrasound Med

Biol, vol. 39, no. 8, pp. 1398-409, Aug 2013, doi:

10.1016/j.ultrasmedbio.2013.02.005.

[22] J. Vidal-Jove, E. Perich, and M. A. Del Castillo, "Ultrasound Guided High

Intensity Focused Ultrasound for malignant tumors: The Spanish experience of

survival advantage in stage III and IV pancreatic cancer," Ultrason Sonochem,

vol. 27, pp. 703-706, Nov 2015, doi: 10.1016/j.ultsonch.2015.05.026.

[23] J. E. Parsons, C. A. Cain, G. D. Abrams, and J. B. Fowlkes, "Pulsed cavitational

ultrasound therapy for controlled tissue homogenization," (in eng), Ultrasound in

Medicine and Biology, vol. 32, no. 1, pp. 115-29, Jan 2006, doi: S0301-

5629(05)00374-1 [pii]

10.1016/j.ultrasmedbio.2005.09.005.

[24] J. E. Parsons, C. A. Cain, and J. B. Fowlkes, "Cost-effective assembly of a basic

fiber-optic hydrophone for measurement of high-amplitude therapeutic ultrasound

fields," J Acoust Soc Am, vol. 119, no. 3, pp. 1432-40, Mar 2006, doi:

10.1121/1.2166708.

[25] H. R. Holmes et al., "Focused ultrasound extraction (FUSE) for the rapid

extraction of DNA from tissue matrices," (in English), Methods Ecol Evol, Oct 20

2020, doi: 10.1111/2041-210x.13505.

Page 274: Determining the Oncological and Immunological Effects of

237

[26] J. Khirallah et al., "Nanoparticle-mediated histotripsy (NMH) using

perfluorohexane 'nanocones'," Phys Med Biol, vol. 64, no. 12, p. 125018, Jun 20

2019, doi: 10.1088/1361-6560/ab207e.

[27] K. C. Longo et al., "Robotically Assisted Sonic Therapy (RAST) for Noninvasive

Hepatic Ablation in a Porcine Model: Mitigation of Body Wall Damage with a

Modified Pulse Sequence," Cardiovasc Intervent Radiol, vol. 42, no. 7, pp. 1016-

1023, Jul 2019, doi: 10.1007/s00270-019-02215-8.

[28] A. R. Smolock et al., "Robotically Assisted Sonic Therapy as a Noninvasive

Nonthermal Ablation Modality: Proof of Concept in a Porcine Liver Model,"

Radiology, vol. 287, no. 2, pp. 485-493, May 2018, doi:

10.1148/radiol.2018171544.

[29] W. W. Roberts, D. Teofilovic, R. C. Jahnke, J. Patri, J. M. Risdahl, and J. A.

Bertolina, "Histotripsy of the prostate using a commercial system in a canine

model," The Journal of urology, vol. 191, no. 3, pp. 860-865, 2014.

[30] M. Shpoliansky, Y. Nevo, Y. Klein, and M. Amitai, "The Accuracy of Early Post-

Operative Abdominal CT Scan," Global Journal of Surgery, vol. 7, no. 1, pp. 19-

25, 2019.

[31] L. C. Sebeke et al., "Feasibility study of MR-guided pancreas ablation using high-

intensity focused ultrasound in a healthy swine model," International Journal of

Hyperthermia, vol. 37, no. 1, pp. 786-798, 2020.

[32] L. Ke et al., "The effect of intra-abdominal hypertension incorporating severe

acute pancreatitis in a porcine model," PloS one, vol. 7, no. 3, p. e33125, 2012.

Page 275: Determining the Oncological and Immunological Effects of

238

[33] J. Otto, M. Afify, U. Jautz, V. Schumpelick, R. Tolba, and A. Schachtrupp,

"Histomorphologic and ultrastructural lesions of the pancreas in a porcine model

of intra-abdominal hypertension," Shock, vol. 33, no. 6, pp. 639-645, 2010.

[34] P. Marchetti, E. H. Finke, A. Gerasimidi-Vazeou, L. Falqui, D. W. Scharp, and P.

E. Lacy, "Automated large-scale isolation, in vitro function and

xenotransplantation of porcine islets of Langerhans," Transplantation, vol. 52, no.

2, pp. 209-213, 1991.

[35] M. Bower, L. Sherwood, Y. Li, and R. Martin, "Irreversible electroporation of the

pancreas: definitive local therapy without systemic effects," Journal of surgical

oncology, vol. 104, no. 1, pp. 22-28, 2011.

[36] C. B. Nahm, S. J. Connor, J. S. Samra, and A. Mittal, "Postoperative pancreatic

fistula: a review of traditional and emerging concepts," Clinical and experimental

gastroenterology, vol. 11, p. 105, 2018.

[37] C. Bassi et al., "The 2016 update of the International Study Group (ISGPS)

definition and grading of postoperative pancreatic fistula: 11 Years After," (in

eng), Surgery, vol. 161, no. 3, pp. 584-591, Mar 2017, doi:

10.1016/j.surg.2016.11.014.

Page 276: Determining the Oncological and Immunological Effects of

239

Chapter 7. Immunological Effects of Histotripsy for Cancer Therapy

Alissa Hendricks-Wenger, Ruby Hutchison, Eli Vlaisavljevich, Irving C Allen

This chapter is excerpted from a manuscript in preparation for submission to Frontiers

of Oncology.

Page 277: Determining the Oncological and Immunological Effects of

240

7.1 Abstract

Cancer is the second leading cause of death worldwide despite major advancements in

diagnosis and therapy over the past century. One of the most debilitating aspects of

cancer is the burden brought on by metastatic disease. Therefore, an ideal treatment

protocol would address not only debulking larger primary tumors but also circulating

tumor cells and distant metastases. To address this need, the use of immune

modulating therapies has become a pillar in the oncology armamentarium. A therapeutic

option that has recently emerged is the use of focal ablation therapies that can destroy

a tumor through various physical or mechanical mechanisms and release a cellular

lysate with the potential to stimulate an immune response. Histotripsy is a non-invasive,

non-ionizing, non-thermal, ultrasound guided ablation technology that has shown

promise over the past decade as a debulking therapy. As histotripsy therapies have

developed, the full picture of the accompanying immune response has revealed a wide

range of immunogenic mechanisms that include DAMP and anti-tumor mediator

release, changes in local cellular immune populations, development of a systemic

immune response, and therapeutic synergism with the inclusion of checkpoint inhibitor

therapies. These studies also suggest that there is a reproducible immune effect from

histotripsy therapies across multiple tumor types. Overall, the effects of histotripsy on

tumors show a positive effect on immunomodulation and clinical outcomes that could be

harnessed to provide systemic benefits to patients.

Page 278: Determining the Oncological and Immunological Effects of

241

7.2 Keywords:

Abscopal Effect, Histotripsy, Focused Ultrasound, Immunomodulation

Page 279: Determining the Oncological and Immunological Effects of

242

7.3 Introduction

Although cancer has plagued mankind throughout history, there still remains a

pressing need to improve treatment options. The core pillars of cancer therapy are

chemotherapy, surgery, radiation, immunotherapy, and ablation. The most common

treatment option for solid tumors remains surgical resection, even though many patients

are not candidates due to tumor size, location, or disease progression (Befeler and Di

Bisceglie, 2002; Chiorean and Coveler, 2015; Hsieh et al., 2017; Armengol et al., 2018).

Tumor ablation modalities have been developed as minimally and non-invasive

adjuvants or alternatives to surgery. These procedures include radiofrequency ablation

(RFA), microwave ablation, cryoablation, irreversible electroporation (IRE), high-

intensity focused ultrasound (HIFU), and histotripsy. While these procedures address

many of the issues with traditional surgery, there are still many areas where these

modalities can be improved.

While the goal of the field of oncology research is to find curative therapies, novel

cancer therapies frequently aim for non-curative endpoints such as increased

progression free survival, increased overall survival, decreased tumor burden, and

improved quality of life. In many cases, the therapeutic goals of local ablation therapy

also include a decrease in tumor burden, enhancement of drug delivery to tumors, and

the modulation of the immune system. While immune system modulation offers the

most promise in terms of long-term patient benefit, it is currently the least predictable

and understood benefit of local tumor ablation therapy. The most observable and

Page 280: Determining the Oncological and Immunological Effects of

243

reliable benefit of local tumor ablation modalities is currently considered to be the

decrease in targeted tumor size (Adam et al., 2020). This can lead to improved patient

outcomes by debulking tumors, which can reduce the patient’s overall tumor burden,

improve local vasculature compression, and reduce tumor nutrient utilization.

7.4 The Role of Ablation in Immunomodulation

Given that one of the most debilitating aspects of cancer is the burden brought on

by metastatic disease, an ideal treatment protocol would address not only debulking

larger or debilitating tumors, but also circulating tumor cells and distant metastases.

Once cancer has spread from its primary site, focal debulking of individual tumors are

less likely to be capable of being a curative treatment option. Therefore, an ideal

debulking therapy would not only reduce the size, or fully eliminate, targeted tumors, but

would also stimulate the immune system to seek and destroy metastatic lesions

throughout the body. This phenomenon has been described as the abscopal effect

(Fig.1). There is increasing evidence that immune system activation and systemic

tumor elimination is critically important for the elimination of micrometastatic lesions

distal to the locally treated tumor, that may not be detectable at the time of treatment or

surgery. Before the abscopal effect was formally defined, it was first noted by physicians

at the turn of the 20th century who reported that local, targeted radiation of the spleen in

leukemia patients would cause changes in the patient’s bone marrow leading to the

remission of disease (Senn, 1903; Capps and Smith, 1907). These clinical findings in

combination with many mouse studies led to the abscopal effect being defined by the

Page 281: Determining the Oncological and Immunological Effects of

244

early 1950’s to encompass the effects to distant organs or cancers after applying

ionizing radiation (Gunz, 1953). This effect was originally thought to only occur for

leukemia, where the radiation of the spleen would reduce or eliminate cancerous cells in

the body (Gunz, 1953; Hotchkiss and Block, 1962). However, over the subsequent

decades, additional reports of solid tumors displaying this effect periodically emerged.

For instance, the reduction of the sizes of distant metastases were reported after the

radiation of a single giant follicular lymphoma or a non-Hodgkin's lymphoma tumor

(Nobler, 1969; Antoniades et al., 1977). However, due to the scarcity of reports, the

abscopal effect in response to ionizing radiation of solid tumors, including renal cell

carcinoma, mammary carcinoma, and neuroblastoma, was frequently dismissed and

remained controversial for decades (Yang et al., 1992; Sanchez-Ortiz et al., 2003;

Demaria et al., 2004; Kaminski et al., 2005). To this day, the abscopal effect associated

with radiation therapy remains controversial (Wang et al., 2020). More recently, the

development of non-ionizing ablation therapies has led to an increase in the number of

abscopal effects reported. Thermal ablations, the first ablation therapies that were

developed and widely accepted, also have reported sporadic cases of a systemic

immune mediated effect where untreated, distal metastases were found to

spontaneously regress (Sanchez-Ortiz et al., 2003; Kondo et al., 2006; Kim et al.,

2008). One of these earlier case studies showed that the treatment of a primary renal

cell carcinoma tumor with RFA caused a spontaneous regression of pulmonary

metastases (Sanchez-Ortiz et al., 2003). On the other hand, as more non-thermal

ablation modalities are being developed pre-clinical studies have had more promising

correlations between focal ablation therapy and the systemic effects (Ringel-Scaia et

Page 282: Determining the Oncological and Immunological Effects of

245

al., 2019; Scheffer et al., 2019; Qu et al., 2020). In general, even though there have

been promising trends that these therapies can offer systemic therapeutic effects for

patients, it should be noted that the reproducibility between patients, tumor types, and

therapy modalities is less than optimal (Kepp et al., 2019).

To achieve an abscopal effect, it is not sufficient to simply release non-specific

markers of tissue damage or cell death, if that were the case surgical resections would

stimulate systemic effects. Instead, there is a need to release specific tumor associated

antigens that allow the immune system to build a targeted response (Fig.1). When

focused, local ablation therapies break up the stromal tissues of a tumor this can create

an increased opportunity for immune cells to physically access the tumor (Dromi et al.,

2009; Lu et al., 2009). As the cancer cells are ablated, there is an increased abundance

of accessible tumor antigens that can be recognized by infiltrating antigen presenting

Figure 7-1 Focal Treatment of Targeted Tumor and Mechanism for Systemic Tumor Control. The right diagram demonstrates the principal of the abscopal effect, where the treatment of one tumor can cause other tumors in the body to shrink or be eliminated to varying degrees due to immune system engagement. The left diagram depicts the simplified mechanism for achieving a systemic effect from focal therapies. DC: Dendritic cell. MHC: Major Histocompatibility Complex. TCR: T cell receptor.

Page 283: Determining the Oncological and Immunological Effects of

246

cells (APC), such as dendritic cells (den Brok et al., 2004; Kim et al., 2008). These

APCs can in turn activate lymphocytes to guide a more precise immune response.

Locally, activation of cytotoxic CD8+ T cells can increase the clearance of any surviving

cancer cells within the area treated with the tumor ablation modality (Ito et al., 2019).

For the strongest systemic immune effects, after ablation, dendritic cells will process the

available tumor associated antigens and migrate to the nearest draining lymph nodes

and the spleen. Once in the lymphoid tissues, the dendritic cells present the antigens

through MHC class I or class II receptors to naïve CD4+ and CD8+ T cells, respectively,

along with co-stimulation of CD28 with CD80/86. Studies that show an increase in tumor

specific lymphocytes in secondary lymphoid organs or in peripheral blood have also

established the correlation between these activated cells and systemic decreases in

tumor burden (Wissniowski et al., 2003; Wu et al., 2004; Widenmeyer et al., 2011;

Leuchte et al., 2020). More recently, additional studies have also defined B cell

activation, the maintenance of plasma cells, and even the generation of tumor specific

antibodies following local tumor ablation and dendritic cell activation (Widenmeyer et al.,

2011; Leuchte et al., 2020). While the activation of T and B lymphocytes is a core

feature of the systemic effects of local tumor ablation therapy, there are many

inconsistencies. For example, a study treating hepatocellular carcinoma with RFA

reported that only 91 of the 178 patients that had an elevated neutrophil-to-lymphocyte

ratio, which is an often-used biomarker of systemic immune system activation,

independently correlated to a significantly increased survival when compared to patients

that did not see the increased ratio (Dan et al., 2013). Similarly, in a prospective study

investigating the long-term effects of RFA after treating secondary liver tumors, it was

Page 284: Determining the Oncological and Immunological Effects of

247

found that only 6 of 49 patients studied had increased antibodies, CD4+ T cells, and/or

CD8+ T cells months after treatment (Widenmeyer et al., 2011). While the exact

mechanisms underlying the inconsistent reports of the abscopal effect and systemic

anti-tumor immune responses are yet to be fully defined, changes in the local tumor

microenvironment have been postulated to significantly impact treatment success.

7.4.1 The Tumor Microenvironment and Tumor Associated Immune Cells

To determine the cause of systemic anti-tumor immunity, and therefore how to

modulate it, there is a need to understand what is occurring within the tumor

microenvironment. Within the heterogeneous cell populations of most tumors, there are

a variety of tumor-associated immune cells. In an ideal case, these leukocytes are

eliminating malignant cells. However, in the case of many cancers, the leukocytes are

reprogrammed, or polarized, to support tumor escape and function to promote

tumorigenesis through the generation of a relatively immunosuppressive tumor

microenvironment. These tumors are often referred to as being immunologically “cold”

due to their lack of inflammation and active immune suppression. With multiple types of

pro-tumor immune cells that produce factors that can improve the tumor niche, it is

difficult for the pro-inflammatory/anti-tumor immune response to overcome this “cold”

environment.

The leukocytes most commonly associated with these anti-inflammatory and tumor

promoting properties include tumor-associated macrophages (TAM), myeloid-derived

suppressor cells (MDSC), and tumor-associated neutrophils (TAN) (Fig.2). TAMs are

Page 285: Determining the Oncological and Immunological Effects of

248

generally defined as CD45+ Ly6C- MHCII+ CD11b+ (Ostuni et al., 2015; Fu et al.,

2020). TAMs are also broken down into subsets based on polarization to the M1

macrophage-like phenotype (pro-inflammatory/anti-tumor) CD68+ CD80/86+ CD11c+

iNOS+ and the M2 macrophage-like phenotype (pro-tumor) CD163+ CD204+ CD206+

Arg1+, with M2-like TAMs being the most immunosuppressive (Wu et al., 2020; Zhou et

al., 2020). It should be noted that while the M1-like and M2-like TAMs do share similar

expression patterns and activities, they are not the same as the traditional M1 and M2

polarized macrophages. MDSCs are generally defined as CD45+ CD11b+ CD11c-

MHCII- as well as CD15+ and CD66b+ in humans (Ko et al., 2009; Rodriguez et al.,

2009; Youn and Gabrilovich, 2010). MDSCs are broken into two main subtypes:

monocytic MDSCs Ly6C+ Ly6G- and granulocytic MDSCs Ly6C- Ly6G+ (Youn and

Gabrilovich, 2010). When generally searching for MDSCs, the GR-1 antibody has been

used given that it binds to both Ly6C and Ly6G (Youn and Gabrilovich, 2010). TANs are

defined as CD45+ CD11b+ Ly6G+ and have been reported to express CD66 and CD15

in human cancers (Fridlender et al., 2009; Hajizadeh et al., 2020; Wu and Zhang, 2020).

It should be noted that the markers that have been established for TANs are

indistinguishable from traditional neutrophils, with the most established phenotypic

difference between the two cell groups being that neutrophils have a half-life of 6-8

hours whereas TANs have a significantly increased lifespan (an additional 12-24 hours)

caused by an inhibition of apoptotic pathways and support from cytokines within the

tumor microenvironment (Wislez et al., 2001; van Raam et al., 2008; Summers et al.,

2010; Trellakis et al., 2011; Hajizadeh et al., 2020). Similar in nomenclature to TAMs,

TANs can also be divided into N1 pro-inflammatory and N2 pro-tumor subtypes,

Page 286: Determining the Oncological and Immunological Effects of

249

however due to limitations in markers there is not an established panel for differentiating

N1 from N2 TANs (Fridlender et al., 2009).

Figure 7-2 Pro-Tumor Immune Cells. CD66, CD66b, and CD15 are human specific markers. Gr-1 represents both Ly6C and Ly6G, therefore does not distinguish the MDSC subtypes, MDSCs are monocytic when Ly6C+ and granulocytic when Ly6G+. Most of the markers available for pro-tumor immune cells are surface receptors and ligands, exceptions listed here include the intracellular protein Arg1 and the transcription factor FOXP3.

Page 287: Determining the Oncological and Immunological Effects of

250

These anti-inflammatory leukocytes are typically distributed throughout the tumor

and margins (Fig.2) and function beyond simply being tumor associated. These

leukocytes are frequently referred to as being pro-tumor given that the range of their

activities can actively promote tumor growth and development. These functions include

reducing the impact of anti-tumor immune responses, recruiting additional tumor

supporting cells, helping the tumor grow by stimulating angiogenesis and secreting

growth factors, and assisting in the epithelial-to-mesenchymal transition (Najafi et al.,

2019; Tesi, 2019; Wu et al., 2019). For TAMs, , this includes producing anti-

inflammatory mediators such as IL-10, Arg1, and TGF-β (Lang et al., 2002; Pang et al.,

2017; Arlauckas et al., 2018), promoting the expression of checkpoint inhibitors to

suppress T cells (Pang et al., 2017; Arlauckas et al., 2018), inducing cancer stem cell

proliferation via IL-6 signaling through STAT3 (Wan et al., 2014), and producing VEGF

to stimulate angiogenesis (Bolat et al., 2006; Hu et al., 2017). These effects are also

seen in TANs and MDSCs, which have both been shown to produce similar profiles of

cytokines, checkpoint inhibitors, and growth factors (Gao et al., 2020; Hajizadeh et al.,

2020; Wu and Zhang, 2020; Mabuchi et al., 2021; Zhou et al., 2021).

In addition to these monocyte and granulocyte derived cells, lymphocytes can also

aid in maintaining this “cold” tumor microenvironment. Regulatory T cells, defined as

CD3+ CD4+ CD25+ FOXP3+, can suppress the adaptive immune response (Tanaka

and Sakaguchi, 2019). While MDSCs, TANs, and TAMs are commonly identified as

tumor promoting cells with specific markers and functions, the role of T regulatory cells

is more situationally specific and their presence and ratio are not necessarily a sign of a

Page 288: Determining the Oncological and Immunological Effects of

251

“cold” tumor microenvironment. In most cases, the presence of a high ratio of T

regulatory cells, known for inhibiting the function of other T cells, is a sign of a “cold”

tumor microenvironment. Clinically, in ovarian carcinoma it has been established that

the recruitment of T regulatory cells into the tumor stroma leads to decreased survival of

patients (Curiel et al., 2004). Additionally, for pancreatic cancer, the T regulatory cells

have been found to aid in the pro-carcinogenic inflammation driven by T helper 17 cells

(Vizio et al., 2012). These poor prognostic correlations in patients is tied to the fact that

T regulatory cells can suppress the function of T helper cells through the production of

immunosuppressive cytokines, including IL-10 and TGF-β (Levings et al., 2002;

Kindlund et al., 2017), secrete perforins and granzymes to directly destroy effector T

cells and B cells (Cao et al., 2007), and express the checkpoint inhibitor CTLA-4 to

suppress APC activity (Kalathil et al., 2013). There have also been reports of T

regulatory cells directly increasing the growth of tumors through the stimulation of

cancer stem cells via the NF-κB-IL6-STAT4 signaling axis (Liu et al., 2021).

On the other hand, there are reports of certain cancers, such as colorectal cancer,

which have worse prognoses for patients that have a reduced presence of T regulatory

cells within their tumors (Salama et al., 2009; Ladoire et al., 2011). A proposed

mechanism for the seemingly contradictory immune promoting function of T regulatory

cells in colorectal cancer is associated with the location of these tumors. In the

gastrointestinal tract, T regulatory cells are primed against the microbiota of the

intestinal space instead of the cancer (Ladoire et al., 2011). This secondary, non-

tumoral target for the immune system allows for the immune response to become more

Page 289: Determining the Oncological and Immunological Effects of

252

robust in the colon, which can in turn destroy the cancerous cells, without being

hampered by the anti-inflammatory control of the tumor microenvironment.

7.4.2 Mechanisms for Shifting the Tumor Microenvironment to Pro-Inflammatory

The largest difference between a “cold” and a “hot” tumor microenvironment is

typically considered to be related to the infiltration of anti-tumor, pro-inflammatory T

cells in “hot” or inflamed tumors (Gajewski, 2015). Checkpoint inhibitors are emerging

as the primary therapeutics used to facilitate the shift in the tumor microenvironment

from “cold” to “hot” or immunologically active (Wilky, 2019). In the case of many

cancers, the checkpoint inhibiting pathways are hijacked to prevent immune cells from

eliminating unhealthy or cancerous cells. The two most heavily studied checkpoint

inhibitor pathways in the tumor ablation field are CTLA-4 to CD80/86, blocking the

normal co-stimulatory role of CD80/86 in T cell activation, and PD-1 to PDL1/2 (Fig.3).

CTLA-4 is a checkpoint inhibitor most commonly associated with antigen presentation in

the lymph nodes that minimizes the proliferation of T cells (Wilky, 2019).

Therapeutically, when CTLA-4 on T cells is blocked with a targeted antibody, there is an

increase in overall T cell proliferation and an increased probability of forming a tumor-

specific immune response. While anti-CTLA-4 is used to increase T cell proliferation,

PD-1 inhibitors decrease T cell inhibition and improve function. Under normal

conditions, PD-1 inhibits the activity of T cells and prevents the targeting of healthy host

cells by T lymphocytes (Wilky, 2019). This pathway in healthy tissues prevents

overzealous immune responses that can drive autoimmunity. However, some tumor

Page 290: Determining the Oncological and Immunological Effects of

253

cells hijack this mechanism and increase the expression of the PD-1 ligands (PD-L1/2),

which artificially reduce the activity of anti-tumor T cells within the tumor

microenvironment. The use of antibodies targeting the PD-1 pathway for cancer therapy

has shown increased levels of infiltrating T cells in tumors that express the PD-L1/2

surface receptors. While PD-1 pathway inhibition has a more tumor-specific therapeutic

effect, and typically fewer side effects compared to CTLA-4 based therapies, there is

still a need to improve the implementation of checkpoint inhibition strategies. This is

especially true in immunologically “cold” tumors where these checkpoint targeted

therapeutics have proven to be minimally effective. Unfortunately, even in cases where

these therapies work there are often significant side effects. For example, the systemic

increase in T cell abundance often leads to autoimmune disease-like symptoms such as

fatigue, nausea, vomiting, diarrhea, arthritis, dermatitis, and myalgia (Arnaud‐Coffin et

al., 2019).

Figure 7-3 The Role of Check Point Inhibitors. Antigen Presenting Cells (APC).

Page 291: Determining the Oncological and Immunological Effects of

254

Beyond the biochemical changes in the tumor microenvironment that work to subvert

immune system detection and elimination, physical barriers that can have significant

detrimental effects on therapeutic approaches are also present. Indeed, many tumors

adopt organ-like structures and extensive fibrosis that build highly complex physical

barriers (Plate, 2011; Jiang et al., 2017). Given that the checkpoint inhibitor’s

mechanism is to improve immune system activation and targeted killing of tumor cells, it

is critical that leukocytes have physical access to the malignant cells. In an attempt aid

the checkpoint inhibitor therapies, ablation modalities have been extensively studied in

an attempt to better understand their ability to modulate the immunological aspects of

the tumor microenvironment. While debulking the tumor and reducing a patient’s overall

tumor burden is the primary goal of focal tumor ablation therapies, improved immune

system access is proving to be an added benefit that can shift the local tumor

microenvironment from “cold” to “hot.”

Ablation therapies with a thermal effect have sporadically seen pro-inflammatory

changes in the tumor microenvironment in the weeks and months following treatment

(Saccomandi et al., 2018; Yin et al., 2018; Minami et al., 2019; Xie et al., 2019).

However, in terms of immunomodulatory success, non-thermal modalities seem to have

more reproducibility, predictability, and improved pre-clinical and clinical success

(Bastianpillai et al., 2015; Beitel-White et al., 2019; Pandit et al., 2019; Ringel-Scaia et

al., 2019). The leading hypothesis regarding differences between modalities theorizes

that while both thermal and non-thermal ablation approaches kill tumor cells, non-

thermal ablation modalities release higher levels of native tumor-specific antigens that

Page 292: Determining the Oncological and Immunological Effects of

255

have not been distorted or denatured by heat. For example, one in vitro study showed

that intact neoantigens are released in significantly higher magnitudes from cells treated

with cryoablation and IRE compared to thermal ablation. Not only were there more

tumor antigens released, these antigens were also more potent at stimulating dendritic

cell antigen presentation and cytotoxic CD8+ T cell activation (Shao et al., 2019).

Activated lymphocytes, either in response to tumor-specific antigens or more generally

in response to innate immune system activation driven by the increase in damage-

associated molecular patterns (DAMPs) further promote the shift from a “cold” to a “hot”

tumor microenvironment (Chandra et al., 2016; Domingo-Musibay et al., 2017; Ringel-

Scaia et al., 2019). Thus, it is becoming clear that non-thermal ablation modalities are

capable of inducing robust local and systemic anti-tumor responses, as either an

independent or an adjuvant therapy with immunotherapeutics, to offer significant

promise beyond just focal tumor debulking.

7.5 Histotripsy: Non-Thermal, Non-Invasive Tumor Ablation

Histotripsy is a non-thermal focused ultrasound therapy that utilizes microsecond

(cavitation-cloud histotripsy, CCH) or millisecond (boiling histotripsy, BH) pulsing

regimens to generate cavitation bubble clouds that leads to precise non-thermal tumor

ablation (Fig.4) (Bader et al., 2019). In addition to histotripsy, other non-thermal focused

ultrasound methods that induce cavitation and do not induce substantial thermal effects

are broadly referred to as mechanical HIFU (mHIFU), as opposed to conventional HIFU

that typically refers to thermal ablation. For the purposes of this study, thermal HIFU will

Page 293: Determining the Oncological and Immunological Effects of

256

be referred to as tHIFU to delineate from mHIFU procedures, which will be the focus of

this review.

In histotripsy, the rapid expansion and collapse of cavitation bubbles ablate tissues

into acellular debris (Vlaisavljevich et al., 2016a; Vlaisavljevich et al., 2017; Smolock et

al., 2018). This non-thermal ablation process is well-established and has resulted in

important hallmarks of histotripsy, including high precision and tissue selectivity

(Vlaisavljevich et al., 2013a; Lin et al., 2014; Vlaisavljevich et al., 2015a). For example,

tissues with a higher Young’s Modulus, such as vasculature and collecting ducts, are

more resistant to damage (Lake et al., 2008; Hall et al., 2009; Hempel et al., 2011;

Vlaisavljevich et al., 2013b; Vlaisavljevich et al., 2014a; Vlaisavljevich et al., 2014b;

Khokhlova et al., 2015; Vlaisavljevich et al., 2015b). Unlike the vast majority of other

ablation therapies, because histotripsy is non-thermal, it is not affected by the heat-sink

Figure 7-4 Histotripsy Schematic. Therapeutic ultrasound transducer positioned outside of the body, focuses ultrasound waves within a targeted tissue generating a cavitation bubble cloud.

Page 294: Determining the Oncological and Immunological Effects of

257

effect, and therefore remains safe and efficacious for use near the vasculature (Thanos

et al., 2008; Pillai et al., 2015). Additional benefits of histotripsy include real-time

imaging feedback with standard imaging (ultrasound, MRI, CT), highly precise-

millimeter accuracy, and the ability to treat tumors of arbitrary sizes and shapes (Hall et

al., 2009; Smolock AR, 2018). After treatment, tissues treated with histotripsy have

shown more rapid dissolution of the ablated tissues compared to other ablation

modalities (Vlaisavljevich et al., 2016a; Smolock AR, 2018). For instance, CCH ablation

of healthy rat livers showed rapid involution of treated volumes, granulation, and growth

of healthy hepatocytes within 28 days with minimal scarring (Vlaisavljevich et al.,

2016a). This healing process is more rapid than what has been reported for other

ablation modalities, such as RFA in humans, which has been reported as causing

“thermal fixation,” where the necrotic mass is still present 14 months post-treatment due

to thermal denaturation of the tissue leaving it resistant to being broken down by normal

pathways (Coad et al., 2003).

In mouse models, histotripsy ablation of hepatic, renal, neuroblastoma, and

melanoma tumors has resulted in significantly increased survival (Schade, 2018;

Worlikar et al., 2019; Eranki et al., 2020; Qu et al., 2020). In addition to debulking

tumors, recent evidence has suggested that histotripsy has the capability to induce a

systemic immune response, as evidenced by the attenuation of metastasis and an

improvement in local and distant disease with combination immunotherapy (Worlikar et

al., 2018; Schade et al., 2019; Qu et al., 2020). This effect has been shown in single

tumor treatments, and has also shown abscopal-like decreases in contralateral tumor

growth in a separate untreated tumor (Schade et al., 2019; Eranki et al., 2020; Qu et al.,

Page 295: Determining the Oncological and Immunological Effects of

258

2020). This effect was found to be modest, but statistically significant with histotripsy

ablation compared to the insignificant trend in mice treated with irradiation and

radiofrequency ablation (Qu et al., 2020). The molecular immunological effects caused

by histotripsy, independently and compared to other ablation modalities, can best be

summarized by the proposed mechanism shown in Figure 5 that is described in the

sections below.

7.6 Immunologic Effects of Histotripsy

The immunological responses to histotripsy ablation through cavitation,

regardless of the sub-therapy being applied, are theoretically similar due to comparable

effects on targeted tissues. Cavitation breaks down tissues, ablating cells into

subcellular fragments and acellular debris (Vlaisavljevich et al., 2016b). To date, there

have been no studies to suggest that mHIFU, BH, nor CCH have any significant

differences in immunologic responses. Therefore, for this review, the immune

responses to these modalities will be reviewed together.

7.6.1 Decreases in Pro-Tumor Immune Cells

Being able to reduce the magnitude of tumor supporting immune cells present

within the tumor microenvironment is pertinent due to the correlation of these cells with

poorer outcomes (Zamarron and Chen, 2011; Fleming et al., 2018; Wu et al., 2019).

There have been multiple studies on thermal and non-thermal ablation modalities that

show either the therapy directly eliminates cells that support the tumor

Page 296: Determining the Oncological and Immunological Effects of

259

microenvironment or the damage initiated by the ablation reprograms the leukocytes

within the tumor microenvironment to shift the microenvironment to one that is more

harsh towards tumor progression (Keisari, 2017; Kepp et al., 2019).

Figure 7-5 Schematic of Histotripsy’s Postulated Roles in Immune System Modulation. Immune processes, cells, and molecules that have been shown to be modulated by focused ultrasound therapies after mechanical ablation are summarized based upon immune system functions: pro-tumor immune cells, cellular immunity, and systemic immunity.

Page 297: Determining the Oncological and Immunological Effects of

260

In the case of histotripsy, there has been one report from Phak et al. demonstrating the

supernatant from cells treated with BH in vitro polarizes naïve macrophages (M0) into

anti-tumor macrophages (M1) and the redifferentiation of tumor associated

macrophages (M2-like phenotype) to the more inflammatory M1 phenotype (Pahk et al.,

2019). In this study, breast cancer cells were treated with BH at varying doses in vitro

and the supernatant collected. Analysis found increased pro-inflammatory signaling

molecules including TNF, which is a potent and well established M1 stimulating cytokine

(Wang and Lin, 2008; Kratochvill et al., 2015). After THP-1 human monocyte cells were

cultured with the BH supernatant they displayed physical and morphological changes

consistent with polarization to M1 macrophages and showed significantly elevated gene

expression consistent with M1 polarization (Pahk et al., 2019). Together, the data

presented in this study suggest that monocytes and macrophages near the histotripsy

ablation region polarize to pro-inflammatory, anti-tumor phenotypes. Likewise, Phak et

al. continued in vitro studies showing that the BH supernatant can also stimulate M2

cells to repolarize to M1, thus leading to a decrease in M2 pro-tumor macrophages

(Fig.5). Thus, these data further suggest that any tumor-associated macrophages

remaining within the treatment zone have the potential to repolarize from an M2-like

state to M1, similar to the cells at the margins of treatment. If this can be recapitulated in

vivo, this would help further decrease the presence of pro-tumor immune cells and

could significantly alter the shift the tumor microenvironment from “cold” to “hot.”

While it might seem safe to assume that any pro-tumor cells within the tumor

microenvironment of treatment zone in vivo would be ablated due to the subcellular

fractionation of histotripsy, there is still a window for any cells that survive from a partial-

Page 298: Determining the Oncological and Immunological Effects of

261

ablation to proliferate and maintain the pro-tumor tumor microenvironment. No literature

confirms this hypothesis for histotripsy ablation in vivo. However, these findings are

consistent with results following RFA, microwave ablation, and IRE, which can all

reduce the presence of tumor supporting immune cells in the tumor microenvironment

(Keisari, 2017; Dai et al., 2021). This dynamic has also been seen in human patients.

For instance, studies of pancreatic adenocarcinoma patients treated with IRE found a

decrease in T regulatory cells in peripheral blood in the days following treatment (Beitel-

White et al., 2019; Pandit et al., 2019). Another study of patients with primary liver

tumors treated with RFA showed a strong correlation between the decrease in MDSCs

and the increase in patients’ survival (Wang et al., 2016). The importance of the

attenuation of pro-tumor immune cells, their decrease in function, and reduction in

magnitude after ablation are all generally correlated with reduced survival in human

patients. Therefore, it is important for the field of histotripsy to further understand the

effects of the ablation on this aspect of the immunomodulatory mechanism.

7.6.2 Histotripsy Produces Damage Associated Molecular Patterns that Directly

Increase Local Cellular Immune Responses

Histotripsy generates subcellular fragments of targeted cells through mechanical

fractionation (Vlaisavljevich et al., 2016b). This, in turn, releases a multitude of damage

associated molecular patterns (DAMPs) that have the potential to stimulate the innate

immune system and significantly alter the tumor microenvironment. DAMPs are

molecules that are found extracellularly following most forms of pathological cell death.

Page 299: Determining the Oncological and Immunological Effects of

262

These molecules are readily identified by the immune system, which in turn elicits a

robust innate immune response. Several DAMPs have been observed and evaluated in

the context of tumor ablation, including the endoplasmic reticulum associated protein

calreticulin (CRT), the non-histone nuclear binding protein HMGB1, the intracellular

energy molecule adenosine 5’-triphosphate (ATP), and various heat shock proteins

(HSP) that were originally identified as DAMPs released after exposure to elevated

temperatures (Harris and Andersson, 2004; Pisetsky, 2011; Korbelik et al., 2015; Chen

et al., 2019; Fodor et al., 2020). Once released from the injured cells or neighboring

cells following damage, these molecules can stimulate a robust immune response by

bindig to receptors known as pattern recognition receptors. As an example, HMGB1

commonly activates the Toll-like Receptors 2 and 4, which classically signals through

NF-κB to drive multiple biological responses, including inflammation and cell death (van

Beijnum et al., 2008).

These DAMPs are emerging as key mediators of the ensuing immune response

following histotripsy. Specifically, following histotripsy treatment of cancer spheroids,

HMGB1, CRT, and HSP were identified in cell supernatants and correlated to levels

expected from within the tumor after an in vivo ablation (Pahk et al., 2019). Additionally,

murine tumors treated with histotripsy have been found to have elevated levels of

HMGB1, CRT, and HSP within the tumor and increased HMGB1 has been routinely

found in serum following treatment (Hu et al., 2005; Schade, 2018; Qu et al., 2020).

DAMPs released in serum function to systemically prime the immune system and can

function to recruit increased circulating leukocytes that can ultimately congregate at the

site of tissue injury following ablation. Together, the local and systemic presence of

Page 300: Determining the Oncological and Immunological Effects of

263

DAMPs aid additional immune cell migration, shift the tumor microenvironment, and

may eventually serve as effective biomarkers of ablation success.

While DAMP release has been confirmed following histotripsy, it is also critical to

determine if these signals are in their naïve and unaltered structural configurations. For

thermal ablation modalities, it has been well established that proteins can be denatured

under high heat settings, making them less efficacious and predictable in stimulating the

immune system. One study investigated the difference in the release of ATP and hsp60

between mHIFU and tHIFU (Hu et al., 2005). This study found that mHIFU is capable of

releasing higher levels of both molecules after treatment, and that the DAMPs released

from mHIFU were more capable of stimulating downstream immunologic changes, such

as activating dendritic cells (Hu et al., 2005). The ability of mHIFU to release a higher

magnitude of active DAMPs compared to tHIFU adds to the hypothesis that non-thermal

ablation therapies are more immunomodulatory compared to thermal modalities.

These data point to histotripsy therapies having the ability to release molecules that

can drive anti-tumor immune system activation, for example by initiating a robust innate

immune response. However, the data associated with the specific DAMPs released

following histotripsy is not consistent across studies. For example, a recent study of

subcutaneous murine neuroblastoma treated with mHIFU showed no significant

changes in HMGB1 levels (Eranki et al., 2020). This could be due to a difference in

tumor types. For example, neuroblastoma did not see the change in DAMPs after

histotripsy that was observed in colon adenocarcinoma, renal cell carcinoma, and

breast adenocarcinoma reported elsewhere (Hu et al., 2005; Schade, 2018; Eranki et

al., 2020; Qu et al., 2020). Alternatively, there are potential differences in histotripsy and

Page 301: Determining the Oncological and Immunological Effects of

264

other mHIFU ablation modalities that are not fully understood and may have played a

role. Another possibility is that differences in DAMP release between a partial ablation

versus a more complete treatment are responsible for these differences. All of these

variables could potentially affect the quality and quantity of DAMPs released. Likewise,

the actual DAMPs generated could significantly vary depending on a number of

experimental factors. Although the aforementioned DAMPs are currently the most

defined following histotripsy, any DAMP could conceivably function as an activator of

the innate immune system after ablation. Future investigation into the effects of other

DAMPs, such as nuclear and mitochondrial DNA, reactive oxygen species, and calcium

ions could prove useful for improving adjuvant therapies (Gong et al., 2019). Additional

studies are needed to determine any potential difference in the quantity and quality of

the release of DAMPs from the different mechanical focused ultrasound therapies.

7.6.3 Pro-Inflammatory Cytokines and Chemokines Associated with Histotripsy Based

Tumor Ablation Modalities

Cytokines and chemokines are critical for cell-to-cell communication and immune

system modulation following damage (Fig. 5). These molecules stimulate the

differentiation and activation of local immune cells and systemically recruit additional

cells to the site of damage. One of the critical cytokines found to be significantly altered

in multiple histotripsy studies is INFγ (Pahk et al., 2019; Schade et al., 2019; Eranki et

al., 2020; Qu et al., 2020). In an in vitro study on breast adenocarcinoma and an in vivo

murine study of neuroblastoma, there was an approximately 2-fold increase in INFγ

Page 302: Determining the Oncological and Immunological Effects of

265

after histotripsy (Pahk et al., 2019; Eranki et al., 2020). However, in an in vivo study

where Eker rat renal cell carcinoma tumors were treated with histotripsy, there was a

significant decrease, ~6pg/mL down to ~2pg/mL, in treated kidneys (Schade et al.,

2019). This decrease was attributed to the native kidney tissues’ defense mechanism

to protect against acute and chronic kidney injuries (Chung and Lan, 2011). The

importance of changes to INFγ is highlighted by its role in activating anti-tumor APCs

and effector cells, inducing ischemia within the tumor by acting on the endothelial cells

thus reducing the stability of intratumoral vasculature, and initiating tumor cell death

through the activation of apoptosis and necroptosis pathways (Ni and Lu, 2018).

However, IFNγ is also a double-edged sword in the tumor microenvironment. In addition

to its function as a potent pro-inflammatory mediator, IFNγ can also up-regulate PD-L1

in some tumors as part of the IRF1 axis, resulting in immune evasion (Mimura et al.,

2018).

While INFγ is the most consistently reported inflammatory mediator across

therapies and tumor types, other important cytokines have been reported as either

significantly or notably upregulated in the days after histotripsy treatment including IL-6,

IL-2, TNF, IL-8, IL-13, and IL-10 (Schade, 2018; Pahk et al., 2019; Eranki et al., 2020;

Qu et al., 2020). While more research is needed to more accurately determine the state

of cytokines in specific tumor types after mechanical ablation, the general picture

appears positive. However, high levels of IL-6, IL-10, and IL-8 in serum have been

reported to negatively correlate with survival in pancreatic cancer patients, due to their

role in stimulating inflammation and adverse changes in the tumor microenvironment

that spurs the growth and development of pancreatic tumors (Feng et al., 2018; Shadhu

Page 303: Determining the Oncological and Immunological Effects of

266

and Xi, 2019). Although this could correlate to a foreboding warning against histotripsy

therapies, at least for pancreatic applications, the induction of these pathways actually

correlates with improved anti-tumor immune responses and increased survival in mice

(Qu et al., 2020). It is certainly possible that these results simply reflect the commonly

promoted differences between mice and humans. However, it is also possible that

additional mechanistic insight may be necessary to better translate the findings from

rodents to human patients.

In addition to the cytokines released there are changes in the serum levels of

multiple growth factors as well. For example, after mHIFU ablation of neuroblastomas,

both an increase in GM-CSF and a decrease in VEGF were found within 24 hours of

treatment (Eranki et al., 2020). Increased levels of GM-CSF are positively correlated

with increased APC cell differentiation and activation. Decreased levels of VEGF are

positively correlated with better clinical outcomes, due to the rate of vascularization of

tumors affecting tumor growth and immunosuppressive effects (Lapeyre-Prost et al.,

2017). The finding that histotripsy can increase GM-CSF and decrease VEGF adds to

the potential for histotripsy to shift the tumor promoting immune microenvironment to

one that is more proinflammatory and tumor suppressive.

Page 304: Determining the Oncological and Immunological Effects of

267

7.6.4 Histotripsy Significantly Alters Immune Cell Populations Systemically and in the

Tumor Microenvironment

With the increased release of DAMPs and anti-tumor mediators, there is a

resultant change in intra-tumoral immune cell populations. In response to damage, the

cells associated with innate immunity are the most rapidly recruited. For histotripsy

ablation, this has been reported to include neutrophils, natural killer cells, dendritic cells,

and macrophages (Fig. 5). In an in vitro experiment, the supernatant of mHIFU treated

murine colon adenocarcinoma cells was shown to activate macrophages (Hu et al.,

2005). This macrophage activation was established through the release of TNF from the

macrophages, which was significantly greater after mHIFU compared to both tHIFU and

controls (Hu et al., 2005). Additionally, in vivo treatment of melanoma tumors with CCH

showed increases in neutrophils, natural killer cells, dendritic cells, and macrophages

10 days after treatment (Qu et al., 2020). This increase in intratumoral immune cell

populations indicate that the tumor microenvironment has become more

immunologically “hot” and these cell populations typically correlate to pro-inflammatory

and anti-tumor changes in the tumor microenvironment.

Beyond innate immune cells, modulation of adaptive immune cells after ablation

has also been strongly correlated with clinical success (Keisari, 2017; Kepp et al.,

2019). In the same melanoma CCH ablation study, increases in T helper cells and B

cells within the treated tumors were observed 10 days post-treatment (Qu et al., 2020).

In addition to the increase in these adaptive immune cell populations, the study also

found a decrease in intratumoral T regulatory cells (Qu et al., 2020). The increase in

Page 305: Determining the Oncological and Immunological Effects of

268

helper T cells and B cells is a further indication that the TME has shifted to a more

permissible, pro-inflammatory environment and further illustrates the improved

accessibility of these cells to the tumor. In general, the influx of leukocytes appears to

reflect the early induction of the innate immune system and eventually yields to cells

associated with a robust adaptive immune response over the course of a few weeks

following treatment. Together, these changes are predicted to have significant impacts

on the local and systemic effects on metastatic lesions. However, it is critical to note

that, while the increased presence of these pro-inflammatory immune cells within the

residual treated tumor does hint at a stronger and more robust overall immune

response, these cells must be specifically primed for tumor antigens found and either in

the circulation or in secondary lymphoid organs to effectively control the overall disease

burden.

7.6.5 Improved Engagement of the Adaptive Immune System and Increases in the

Systemic Anti-Tumor Immune Response

The initiation of the systemic anti-tumor immune response requires the

generation of both high quantity and high-quality tumor antigens. Previous studies with

thermal ablation, cryoablation, and IRE established that the non-thermal ablation

modalities appear to release antigens that are significantly better at driving predictable

and effective antigen presentation (Shao et al., 2019). This ultimately results in

improved systemic immune responses (Shao et al., 2019). Although an explicit study

has yet to explore if histotripsy ablation negatively impacts the quality of released

Page 306: Determining the Oncological and Immunological Effects of

269

antigens, studies have been conducted that indirectly evaluate this mechanism. A study

using the B16F10 murine melanoma cell line and a transgenic variant of the cell line

revealed an increase in the immunomodulatory effects of histotripsy in the tumors that

expressed an immunogenically active antigen (Qu et al., 2020). In this study, only mice

that had tumors transfected with the potential antigen GP33, a known antigen from

lymphocytic choriomeningitis virus, and were also treated with histotripsy, generated

CD8+ T cells that were capable of producing IFNγ after stimulation with IL-2 and

brefeldin A. These two agents stimulate memory CD8+ T cells. Together, these data

imply that transfected antigens are still viable after histotripsy treatment and are able to

stimulate an adaptive immune response.

The activation and migration of dendritic cells to tumor-draining lymph nodes and

the spleen are further evidence of systemic immune system activation. Early studies

into dendritic cells found the supernatants collected from in vitro mHIFU treatments

were able to stimulate immature dendritic cells to express higher levels of CD80 and

CD86, which are markers of activation, and enhanced secretion of IL-12, which are

utilized by dendritic cells to activate both CD8+ cytotoxic and CD4+ helper T cells

(Watford et al., 2003; Hu et al., 2005; Henry et al., 2008). Additional murine studies with

various tumor types further established the migration of, or the increase in, the number

of dendritic cells to the tumor-draining lymph nodes and spleen (Hu et al., 2007; Huang

et al., 2012; Eranki et al., 2020). The higher presence of this critical antigen-presenting

cell indicates that a subsequent increase in lymphocyte populations is likely.

Once the APCs have been activated, antigens are presented CD8+ and CD4+ T

cells that proliferate and are subsequently recruited back to the treated tumor, as well

Page 307: Determining the Oncological and Immunological Effects of

270

as, circulate systemically to target and kill distal tumor cells expressing the targeted

tumor specific antigens. As discussed earlier, the reduction of T regulatory cells is

important for changing the tumor microenvironment and shifting the immune response

from “cold” to “hot”, allowing these anti-tumor CD4+ and CD8+ T cells access to

malignant cells. Histotripsy has been shown to reduce the magnitude of T regulatory

cells and increase the ratio of CD8+ to T regulatory cells in both the tumor-draining

lymph nodes and spleens of treated mice (Huang et al., 2012; Qu et al., 2020). For

example, the ratio of CD8+:CD4+ cells in the spleen was found to increase after treating

melanoma tumors with CCH (Qu et al., 2020). Additionally, after treating prostate

tumors with mHIFU, the ratio of CD8+:CD4+ cells in the spleen was also found to be

increased, and protective against subsequent tumor challenges (Huang et al., 2012).

However, in treating neuroblastoma tumors with mHIFU, the ratio of CD8+:CD4+ cells in

the spleen was found to decrease (Eranki et al., 2020). This difference in the increase

versus decrease of the CD8+:CD4+ ratio is not necessarily a sign that regulatory T cells

are predominate nor is it indication of poor prognosis. This shift in ratio could indicate

helper T cells primed to increase an immune response in response to the damage that

could lead to a pro-inflammatory response. Beyond regulatory T cells, histotripsy has

been found to increase the number of CD8+ and CD4+ helper cells in the spleen (Xing

et al., 2008; Eranki et al., 2020; Qu et al., 2020). These changes in T cell populations in

secondary lymphoid organs after histotripsy treatment implies that similar changes

occur within the tumor, and that these cells are primed against tumor specific antigens.

While all of these studies established correlative changes in T cells after

histotripsy treatment, it is important to determine the efficacy and tumor specificity of

Page 308: Determining the Oncological and Immunological Effects of

271

programmed cells. CD8+ cells isolated from the spleens of mice that had subcutaneous

colon adenocarcinoma tumors 10 days post-treatment with mHIFU were co-cultured

with the corresponding cell line (Hu et al., 2007). The CD8+ T cells were found to be

more cytotoxicity against the cancer cells compared to the lymphocytes isolated from

mice treated with tHIFU. In this same study, ELIspot assays showed that mHIFU

generated a higher magnitude of tumor-specific CD8+ cells than tHIFU (Hu et al., 2007).

In a similar study with histotripsy treated prostate tumors, CD8+ T cells harvested from

the spleen were found to be tumor-specific and were subsequently activated when

challenged in vitro with the tumor cells (Huang et al., 2012). Finally, in a third study, the

treatment of a known antigen into a cell line before tumor engraftment allowed for the

determination of CD8+ T cells in the tumor-draining lymph nodes that were primed

against tumor-specific antigens after treatment with CCH (Qu et al., 2020). Together,

these studies support the hypothesis that histotripsy treatment of the local tumor is

effective at generating tumor specific CD8+ cytotoxic T cells that are found systemically

in distal lymph nodes and the spleen.

The efficacy of adaptive immune system activation is shown through systemic

tumor killing both through the reduction of metastasis and control of contralateral

tumors. Several studies have evaluated this phenomenon. For example, in a study

using immunocompetent New Zealand White rabbits with VX-2 (leporine papilloma)

tumors grown within a single kidney, histotripsy treatment on day 13 did not show a

change in metastasis by day 19 (Styn et al., 2012). Ultimately, the authors associated

the lack of difference to the aggression of the cancer (Styn et al., 2012). Beyond this

study, it should be noted that the majority of histotripsy studies have actually shown

Page 309: Determining the Oncological and Immunological Effects of

272

significant improvements in metastasis (Xing et al., 2008; Qu et al., 2020). For example,

in a murine melanoma model, tumors were grown subcutaneously, treated with

histotripsy, and amputated two days after treatment (Xing et al., 2008). For most studies

conducted to date, decreases in lung metastasis are correlated to systemic cancer

control and suggestive of an abscopal response. However, as an alternative

interpretation of the data, the decrease in metastasis could simply be an effect of the

primary tumor ablation not being able to produce as many circulating tumor cells due to

its reduced size. This is a common critique of focal tumor ablation studies that report

changes in metastatic burden.

Based on the current data in the field, we believe that it is likely that both

interpretations are accurate; whereby, the decrease in metastasis is due to both the

activation of the systemic anti-tumor immune response and the debulking of the primary

tumor. Thus, it is essential that future mechanistic work in the field design experiments

designed to better establish causation. For example, in addition to the prevention of

additional tumor growth through reduced metastasis, there are multiple reports of CD8+

cytotoxic T cells in contralateral tumors (Schade et al., 2019; Qu et al., 2020). To

evaluate histotripsy in these contralateral models, mice were inoculated with bilateral

subcutaneous B16GP33 melanoma tumors and after 10 days of tumor growth one of

these tumors were treated. In the treated tumor, there was a sharp infiltration of CD8+ T

cells that localized in the ablation margin, while over the course of a week the

contralateral-untreated tumor slowly saw a perfuse increase in CD8+ T cells (Qu et al.,

2020). This study establishes that in injected, identical tumors that a systemic effect can

be achieved. As a de novo model of contralateral tumors, Eker rats have been

Page 310: Determining the Oncological and Immunological Effects of

273

deployed. Eker rats are a model with an insertion in the rat homologue of the human

tuberous sclerosis gene (TSC2) that spontaneously develop multiple renal cell

carcinoma tumors throughout both of their kidneys (Cook and Walker, 2004). In a study

of boiling histotripsy, Eker rats had one approximately 0.5cm3 tumor treated and after

48hrs, their treated and contralateral kidneys were collected for immunohistochemistry

staining for CD8+ cells. This found that if one tumor was treated then both tumor riddled

kidneys would see an influx of CD8+ T cells, while sham treated rats did not see a

change in either kidney (Schade et al., 2019). These contralateral tumors have been

reported to have decreased growth rates after histotripsy treatment of the targeted

primary tumor, and more significantly than radiation ablation or RFA (Qu et al., 2020).

Using mice with B16GP33 tumors, after 10 days of tumor growth mice were either

treated with histotripsy, RFA, radiation therapy, or sham-control treated. This study

found that the mice that were treated with histotripsy saw a significant increase in tumor

infiltrating CD8+ T cells in treated tumors compared to RFA and radiation ablation,

neither of which saw any increase from sham-controls (Qu et al., 2020). This not only

further strengthens the idea that histotripsy can stimulate a systemic immune response,

but also supports the idea that non-thermal and non-ionizing therapies are more

effective.

While there is a reduction in metastasis in these studies, the more established

contralateral tumors only have their growth slowed, not inhibited nor reversed. This

implies that while there is a systemic immune response against the cancer, it is not

strong enough to prevent growth or be curative without combination with other

therapeutic methods or additional treatments. Further, the efficacy of mHIFU treatment

Page 311: Determining the Oncological and Immunological Effects of

274

of generating systemic tumor-specific protection has been shown with the reduction in

tumor growth rate in challenge tumors injected 6 days after treatment (Hu et al., 2007).

Notably, the effects of mHIFU to minimize the growth rates of treated tumors were more

effective than tHIFU, even though the debulking on the primary tumor was much greater

(83% and 43% reduction in tumor volume respectively) (Hu et al., 2007). This indicates

that the non-thermal ablation modality stimulated a stronger immune response.

However, as with the pre-existing contralateral tumors, the post-treatment injection

challenge still allowed for tumor growth. Together these studies reveal that histotripsy

has the power to prime the immune system sufficiently to reduce systemic tumor burden

through control of naturally occurring metastasis, spontaneous similar-primary tumors,

and secondary tumors injected as a challenge.

7.6.6 Combination Therapy with Checkpoint-Inhibitors

Given that many cancers are immunologically resistant to checkpoint-inhibitors

due, in part, to the immunological state of the tumor microenvironment, it is critical to

explore the potential of histotripsy as a strategy to improve tumor responsiveness. The

increased release of DAMPs, shift in the inflammatory state of the tumor

microenvironment, enhanced recruitment of immune cells within the tumor, and

evidence of a systemic anti-tumor immune response all discussed above indicate that

histotripsy can have local and systemic immunomodulatory effects that may be

favorable for these classes of therapeutics. Based on these changes, it is reasonable to

hypothesize that the shift of the tumor microenvironment to a “hot” environment should

Page 312: Determining the Oncological and Immunological Effects of

275

increase the effects of checkpoint inhibitors allowing for a potentially stronger anti-tumor

immune response. The efficacy of anti-CTLA-4, anti-PD-1, and anti-PD-L1 therapies is

correlative to the levels of expression of CTLA-4, CD80/86, PD-1, and PD-L1 in the

patients’ tumor cells, APCs, and T cells (Fig.3). Therefore, before studying the effects of

specific checkpoint inhibitors, it is critical to determine if there is supporting data to

suggest that histotripsy may have a synergistic effect (Wilky, 2019). For example, anti-

PD-L1 targeted therapies are not typically used in cases where tumors do not express

high levels of PD-L1. However, this is not always a clear decision, especially for

checkpoint inhibitors that can be induced or up-regulated following treatment. Take for

instance the expression of CD80/86 on dendritic cells, which was found to increase in

vitro when stimulated by mHIFU supernatant, indicating that the lysate released from

cells after histotripsy treatment can stimulate an upregulation of a checkpoint inhibitor

(Hu et al., 2005). In vivo, CD86 was significantly increased on dendritic cells found in

the spleen, and CD80 was increased significantly on dendritic cells in both spleen and

tumor draining lymph nodes (Huang et al., 2012). It has also been shown that there is

increase in PD-L1 in tumors 72 hours after treatment with mHIFU (Eranki et al., 2020).

As mentioned above, this could be due to the downstream effects of IFNγ production

following treatment. While this is typically detrimental to the systemic immune response,

these in vivo upregulations of checkpoint molecules after histotripsy treatment actually

indicates that there may be an increased window of opportunity to utilize checkpoint

inhibitors for synergistic therapy to potentially improve anti-tumor immunity.

With the multiple studies suggesting that histotripsy ablation can increase the

expression of both CTLA-4 and PD-1 pathway receptors, it is reasonable to hypothesize

Page 313: Determining the Oncological and Immunological Effects of

276

that histotripsy in combination with checkpoint inhibitor therapies should have a

synergistic effect. This has been explored in a pair of recent studies. In one study,

individual CTLA-4 antibody therapy and CCH ablation decreased the rate of

contralateral tumor growth, but there was an even more significant decrease with

combined therapy in both melanoma and hepatocellular carcinoma (Qu et al., 2020).

For this study, one group of mice bearing subcutaneous B16GP33 melanoma tumors

were administered two doses of CTLA-4 monoclonal antibody prior to histotripsy and an

additional treatment afterwards. Another group of mice bearing subcutaneous Hepa1-6

hepatocellular carcinoma tumors were given three doses of CTLA-4 monoclonal

antibody prior and one post histotripsy treatment. For both of these tumor studies,

melanoma and hepatocellular carcinoma, the combination of CTLA-4 and histotripsy

yielded significantly reduced tumor volume compared to either treatment individually

(Qu et al., 2020). In a second study, three doses of both CTLA-4 and PD-L1 antibodies

in the days following mHIFU treatment of subcutaneous neuroblastoma tumors

significantly increased the survival of animals compared to other therapeutic

combinations (Eranki et al., 2020). This study went further and performed a similar

experiment, but instead of only looking at the effects of a single mHIFU treated tumor

there was a second tumor grown contralaterally. In this report, after unilateral treatment

with mHIFU and systemic CTLA-4 and PD-L1 therapy there was complete remission of

both tumors, an effect that was not seen with any other therapy combination (Eranki et

al., 2020). Together, these studies demonstrate the improved therapeutic effects of the

combined therapy over either histotripsy or checkpoint inhibitors independently.

Page 314: Determining the Oncological and Immunological Effects of

277

7.7 Conclusion and Future Outlook

As histotripsy therapies have been developed over the past decade, the

knowledge about the immune response has started to develop a more complete picture

about the mechanisms of action from DAMP and anti-tumor mediator release, to

changes in local cellular immune populations, development of a systemic immune

response, and therapeutic synergism with the inclusion of checkpoint inhibitor therapies

(Fig. 5). In sum, these studies suggest that there is a reproducible, consistent, and

perhaps even tunable immune effect generated by histotripsy modalities that is

consistent across multiple tumor types. However, while these data are certainly exciting

and potentially paradigm shifting, the number of studies in this field are still quite limited

and most are based on murine models with inconsistent tumor ablation quality. Looking

towards the future, there needs to be an effort to compare the various histotripsy

treatments and doses, as well as other mHIFU methods, in order to better understand

the relationship between the extent of ablation in stimulating an immune response.

Likewise, as these therapies begin to be utilized in human trials, it will be crucial to

translate these findings into actionable results relevant to human patients. Additional

basic studies and preclinical animal trials are also still needed to develop missing

mechanistic insight and more translationally relevant studies are needed to ensure

these findings occur outside of the typical model organisms. Despite these limitations,

the benefits of histotripsy over other thermal ablation modalities suggest the potential of

this focal tumor ablation therapy to induce a systemic anti-tumor immune response and

Page 315: Determining the Oncological and Immunological Effects of

278

therefore supports the hypothesis that histotripsy has a high likelihood of positively

impacting the clinical outcomes for cancer patients.

7.8 Acknowledgements

This work was supported by the Virginia Maryland College of Veterinary Medicine;

The Virginia Tech Institute for Critical Technology and Applied Sciences Center for

Engineered Health; the Focused Ultrasound Foundation; and The National Institutes of

Health. The content is solely the responsibility of the authors and does not necessarily

represent the official views of the NIH or any other funding agency. All figures were

created using Biorender.com.

Page 316: Determining the Oncological and Immunological Effects of

279

7.9 References

Adam, R., Kitano, Y., Abdelrafee, A., Allard, M.-A., and Baba, H. (2020). Debulking

surgery for colorectal liver metastases: Foolish or chance? Surgical Oncology 33,

266-269.

Antoniades, J., Brady, L.W., and Lightfoot, D.A. (1977). Lymphangiographic

demonstration of the abscopal effect in patients with malignant lymphomas.

International Journal of Radiation Oncology* Biology* Physics 2(1-2), 141-147.

Arlauckas, S.P., Garren, S.B., Garris, C.S., Kohler, R.H., Oh, J., Pittet, M.J., et al.

(2018). Arg1 expression defines immunosuppressive subsets of tumor-

associated macrophages. Theranostics 8(21), 5842.

Armengol, C., Sarrias, M.R., and Sala, M. (2018). Hepatocellular carcinoma: present

and future. Medicina Clínica (English Edition) 150(10), 390-397.

Arnaud‐Coffin, P., Maillet, D., Gan, H.K., Stelmes, J.J., You, B., Dalle, S., et al. (2019).

A systematic review of adverse events in randomized trials assessing immune

checkpoint inhibitors. International Journal of Cancer 145(3), 639-648.

Bader, K.B., Vlaisavljevich, E., and Maxwell, A.D. (2019). For whom the bubble grows:

physical principles of bubble nucleation and dynamics in histotripsy ultrasound

therapy. Ultrasound in medicine & biology.

Bastianpillai, C., Petrides, N., Shah, T., Guillaumier, S., Ahmed, H.U., and Arya, M.

(2015). Harnessing the immunomodulatory effect of thermal and non-thermal

ablative therapies for cancer treatment. Tumor Biology 36(12), 9137-9146.

Page 317: Determining the Oncological and Immunological Effects of

280

Befeler, A.S., and Di Bisceglie, A.M. (2002). Hepatocellular carcinoma: diagnosis and

treatment. Gastroenterology 122(6), 1609-1619.

Beitel-White, N., Martin, R.C.G., Li, Y., Brock, R.M., Allen, I.C., and Davalos, R.V.

(2019). Real-time prediction of patient immune cell modulation during irreversible

electroporation therapy. Sci Rep 9(1), 17739. doi: 10.1038/s41598-019-53974-w.

Bolat, F., Kayaselcuk, F., Nursal, T., Yagmurdur, M., Bal, N., and Demirhan, B. (2006).

Microvessel density, VEGF expression, and tumor-associated macrophages in

breast tumors: correlations with prognostic parameters. Vascular 14, 15.

Cao, X., Cai, S.F., Fehniger, T.A., Song, J., Collins, L.I., Piwnica-Worms, D.R., et al.

(2007). Granzyme B and perforin are important for regulatory T cell-mediated

suppression of tumor clearance. Immunity 27(4), 635-646.

Capps, J., and Smith, J. (1907). Experiments on the Leukolytic Action of the Blood

Serum of Cases of Leukaemia Treated with X-Ray and the Injection of Human

Leukolytic Serum in a Case of Leukaemia. The Journal of experimental medicine

9(1), 51-63.

Chandra, D., Jahangir, A., Cornelis, F., Rombauts, K., Meheus, L., Jorcyk, C.L., et al.

(2016). Cryoablation and Meriva have strong therapeutic effect on triple-negative

breast cancer. Oncoimmunology 5(1), e1049802. doi:

10.1080/2162402x.2015.1049802.

Chen, F., Bao, H., Xie, H., Tian, G., and Jiang, T. (2019). Heat shock protein expression

and autophagy after incomplete thermal ablation and their correlation.

International Journal of Hyperthermia 36(1), 95-103.

Page 318: Determining the Oncological and Immunological Effects of

281

Chiorean, E.G., and Coveler, A.L. (2015). Pancreatic cancer: optimizing treatment

options, new, and emerging targeted therapies. Drug Des Devel Ther 9, 3529-

3545. doi: 10.2147/DDDT.S60328.

Chung, A.C., and Lan, H.Y. (2011). Chemokines in renal injury. Journal of the American

Society of Nephrology 22(5), 802-809.

Coad, J.E., Kosari, K., Humar, A., and Sielaff, T.D. (2003). Radiofrequency ablation

causes ‘thermal fixation’of hepatocellular carcinoma: a post‐liver transplant

histopathologic study. Clinical transplantation 17(4), 377-384.

Cook, J.D., and Walker, C.L. (2004). The Eker rat: establishing a genetic paradigm

linking renal cell carcinoma and uterine leiomyoma. Curr Mol Med 4(8), 813-824.

doi: 10.2174/1566524043359656.

Curiel, T.J., Coukos, G., Zou, L., Alvarez, X., Cheng, P., Mottram, P., et al. (2004).

Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune

privilege and predicts reduced survival. Nature medicine 10(9), 942-949.

Dai, Z., Wang, Z., Lei, K., Liao, J., Peng, Z., Lin, M., et al. (2021). Irreversible

electroporation induces CD8+ T cell immune response against post-ablation

hepatocellular carcinoma growth. Cancer Letters. doi:

https://doi.org/10.1016/j.canlet.2021.01.001.

Dan, J., Zhang, Y., Peng, Z., Huang, J., Gao, H., Xu, L., et al. (2013). Postoperative

neutrophil-to-lymphocyte ratio change predicts survival of patients with small

hepatocellular carcinoma undergoing radiofrequency ablation. PloS one 8(3),

e58184.

Page 319: Determining the Oncological and Immunological Effects of

282

Demaria, S., Ng, B., Devitt, M.L., Babb, J.S., Kawashima, N., Liebes, L., et al. (2004).

Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is

immune mediated. Int J Radiat Oncol Biol Phys 58(3), 862-870. doi:

10.1016/j.ijrobp.2003.09.012.

den Brok, M.H., Sutmuller, R.P., van der Voort, R., Bennink, E.J., Figdor, C.G., Ruers,

T.J., et al. (2004). In situ tumor ablation creates an antigen source for the

generation of antitumor immunity. Cancer research 64(11), 4024-4029.

Domingo-Musibay, E., Heun, J.M., Nevala, W.K., Callstrom, M., Atwell, T., Galanis, E.,

et al. (2017). Endogenous Heat-Shock Protein Induction with or Without

Radiofrequency Ablation or Cryoablation in Patients with Stage IV Melanoma.

Oncologist 22(9), 1026-e1093. doi: 10.1634/theoncologist.2017-0060.

Dromi, S.A., Walsh, M.P., Herby, S., Traughber, B., Xie, J., Sharma, K.V., et al. (2009).

Radiofrequency ablation induces antigen-presenting cell infiltration and

amplification of weak tumor-induced immunity. Radiology 251(1), 58-66.

Eranki, A., Srinivasan, P., Ries, M., Kim, A., Lazarski, C.A., Rossi, C.T., et al. (2020).

High-Intensity Focused Ultrasound (HIFU) Triggers Immune Sensitization of

Refractory Murine Neuroblastoma to Checkpoint Inhibitor Therapy. Clinical

Cancer Research 26(5), 1152-1161.

Feng, L., Qi, Q., Wang, P., Chen, H., Chen, Z., Meng, Z., et al. (2018). Serum levels of

IL-6, IL-8, and IL-10 are indicators of prognosis in pancreatic cancer. Journal of

International Medical Research 46(12), 5228-5236.

Page 320: Determining the Oncological and Immunological Effects of

283

Fleming, V., Hu, X., Weber, R., Nagibin, V., Groth, C., Altevogt, P., et al. (2018).

Targeting myeloid-derived suppressor cells to bypass tumor-induced

immunosuppression. Frontiers in immunology 9, 398.

Fodor, P., White, B., and Khan, R. (2020). Inflammation-The role of ATP in pre-

eclampsia. Microcirculation 27(1), e12585. doi: 10.1111/micc.12585.

Fridlender, Z.G., Sun, J., Kim, S., Kapoor, V., Cheng, G., Ling, L., et al. (2009).

Polarization of tumor-associated neutrophil phenotype by TGF-β:“N1” versus

“N2” TAN. Cancer cell 16(3), 183-194.

Fu, L.-Q., Du, W.-L., Cai, M.-H., Yao, J.-Y., Zhao, Y.-Y., and Mou, X.-Z. (2020). The

roles of tumor-associated macrophages in tumor angiogenesis and metastasis.

Cellular Immunology, 104119.

Gajewski, T.F. (Year). "The next hurdle in cancer immunotherapy: Overcoming the non–

T-cell–inflamed tumor microenvironment", in: Seminars in oncology: Elsevier),

663-671.

Gao, X., Sui, H., Zhao, S., Gao, X., Su, Y., and Qu, P. (2020). Immunotherapy

Targeting Myeloid-Derived Suppressor Cells (MDSCs) in Tumor

Microenvironment. Front Immunol 11, 585214. doi: 10.3389/fimmu.2020.585214.

Gong, T., Liu, L., Jiang, W., and Zhou, R. (2019). DAMP-sensing receptors in sterile

inflammation and inflammatory diseases. Nature Reviews Immunology, 1-18.

Gunz, F. (1953). Bone Marrow Changes in Patients with Chronic Leukemia Treated by

Splenic X-irradiation: Preliminary Report. Blood 8(8), 687-692.

Hajizadeh, F., Maleki, L.A., Alexander, M., Mikhailova, M.V., Masjedi, A., Ahmadpour,

M., et al. (2020). Tumor-associated neutrophils as new players in

Page 321: Determining the Oncological and Immunological Effects of

284

immunosuppressive process of the tumor microenvironment in breast cancer.

Life sciences, 118699.

Hall, T.L., Hempel, C.R., Wojno, K., Xu, Z., Cain, C.A., and Roberts, W.W. (2009).

Histotripsy of the prostate: dose effects in a chronic canine model. Urology 74(4),

932-937.

Harris, H.E., and Andersson, U. (2004). Mini‐review: the nuclear protein HMGB1 as a

proinflammatory mediator. European journal of immunology 34(6), 1503-1512.

Hempel, C.R., Hall, T.L., Cain, C.A., Fowlkes, J.B., Xu, Z., and Roberts, W.W. (2011).

Histotripsy fractionation of prostate tissue: local effects and systemic response in

a canine model. The Journal of urology 185(4), 1484-1489.

Henry, C.J., Ornelles, D.A., Mitchell, L.M., Brzoza-Lewis, K.L., and Hiltbold, E.M.

(2008). IL-12 produced by dendritic cells augments CD8+ T cell activation

through the production of the chemokines CCL1 and CCL17. J Immunol 181(12),

8576-8584. doi: 10.4049/jimmunol.181.12.8576.

Hotchkiss, D.J., and Block, M.H. (1962). Effect of splenic irradiation on systemic

hematopoiesis. Archives of internal medicine 109(6), 695-711.

Hsieh, J.J., Purdue, M.P., Signoretti, S., Swanton, C., Albiges, L., Schmidinger, M., et

al. (2017). Renal cell carcinoma. Nature reviews Disease primers 3, 17009.

Hu, J.M., Liu, K., Liu, J.H., Jiang, X.L., Wang, X.L., Yang, L., et al. (2017). The

increased number of tumor-associated macrophage is associated with

overexpression of VEGF-C, plays an important role in Kazakh ESCC invasion

and metastasis. Experimental and molecular pathology 102(1), 15-21.

Page 322: Determining the Oncological and Immunological Effects of

285

Hu, Z., Yang, X.Y., Liu, Y., Morse, M.A., Lyerly, H.K., Clay, T.M., et al. (2005). Release

of endogenous danger signals from HIFU-treated tumor cells and their

stimulatory effects on APCs. Biochemical and biophysical research

communications 335(1), 124-131.

Hu, Z., Yang, X.Y., Liu, Y., Sankin, G.N., Pua, E.C., Morse, M.A., et al. (2007).

Investigation of HIFU-induced anti-tumor immunity in a murine tumor model.

Journal of translational medicine 5(1), 34.

Huang, X., Yuan, F., Liang, M., Lo, H.-W., Shinohara, M.L., Robertson, C., et al. (2012).

M-HIFU inhibits tumor growth, suppresses STAT3 activity and enhances tumor

specific immunity in a transplant tumor model of prostate cancer. PloS one 7(7),

e41632.

Ito, F., Vardam, T.D., Appenheimer, M.M., Eng, K.H., Gollnick, S.O., Muhitch, J.B., et al.

(2019). In situ thermal ablation augments antitumor efficacy of adoptive T cell

therapy. International Journal of Hyperthermia 36(sup1), 22-36.

Jiang, H., Hegde, S., and DeNardo, D.G. (2017). Tumor-associated fibrosis as a

regulator of tumor immunity and response to immunotherapy. Cancer

Immunology, Immunotherapy 66(8), 1037-1048.

Kalathil, S., Lugade, A.A., Miller, A., Iyer, R., and Thanavala, Y. (2013). Higher

frequencies of GARP+ CTLA-4+ Foxp3+ T regulatory cells and myeloid-derived

suppressor cells in hepatocellular carcinoma patients are associated with

impaired T-cell functionality. Cancer research 73(8), 2435-2444.

Page 323: Determining the Oncological and Immunological Effects of

286

Kaminski, J.M., Shinohara, E., Summers, J.B., Niermann, K.J., Morimoto, A., and

Brousal, J. (2005). The controversial abscopal effect. Cancer treatment reviews

31(3), 159-172.

Keisari, Y. (2017). Tumor abolition and antitumor immunostimulation by physico-

chemical tumor ablation. Front Biosci (Landmark Ed) 22, 310-347.

Kepp, O., Marabelle, A., Zitvogel, L., and Kroemer, G. (2019). Oncolysis without

viruses—inducing systemic anticancer immune responses with local therapies.

Nature Reviews Clinical Oncology, 1-16.

Khokhlova, V.A., Fowlkes, J.B., Roberts, W.W., Schade, G.R., Xu, Z., Khokhlova, T.D.,

et al. (2015). Histotripsy methods in mechanical disintegration of tissue: Towards

clinical applications. International journal of hyperthermia 31(2), 145-162.

Kim, H., Park, B.K., and Kim, C.K. (2008). Spontaneous regression of pulmonary and

adrenal metastases following percutaneous radiofrequency ablation of a

recurrent renal cell carcinoma. Korean journal of radiology 9(5), 470.

Kindlund, B., Sjöling, Å., Yakkala, C., Adamsson, J., Janzon, A., Hansson, L.E., et al.

(2017). CD4(+) regulatory T cells in gastric cancer mucosa are proliferating and

express high levels of IL-10 but little TGF-β. Gastric Cancer 20(1), 116-125. doi:

10.1007/s10120-015-0591-z.

Ko, J.S., Zea, A.H., Rini, B.I., Ireland, J.L., Elson, P., Cohen, P., et al. (2009). Sunitinib

mediates reversal of myeloid-derived suppressor cell accumulation in renal cell

carcinoma patients. Clin Cancer Res 15(6), 2148-2157. doi: 10.1158/1078-

0432.Ccr-08-1332.

Page 324: Determining the Oncological and Immunological Effects of

287

Kondo, S., Okusaka, T., Ueno, H., Ikeda, M., and Morizane, C. (2006). Spontaneous

regression of hepatocellular carcinoma. International journal of clinical oncology

11(5), 407-411.

Korbelik, M., Banáth, J., Saw, K.M., Zhang, W., and Čiplys, E. (2015). Calreticulin as

cancer treatment adjuvant: combination with photodynamic therapy and

photodynamic therapy-generated vaccines. Frontiers in oncology 5, 15.

Kratochvill, F., Neale, G., Haverkamp, J.M., Van de Velde, L.-A., Smith, A.M.,

Kawauchi, D., et al. (2015). TNF counterbalances the emergence of M2 tumor

macrophages. Cell reports 12(11), 1902-1914.

Ladoire, S., Martin, F., and Ghiringhelli, F. (2011). Prognostic role of FOXP3+ regulatory

T cells infiltrating human carcinomas: the paradox of colorectal cancer. Cancer

Immunology, Immunotherapy 60(7), 909-918.

Lake, A., Xu, Z., Wilkinson, J., Cain, C., and Roberts, W. (2008). Renal ablation by

histotripsy—does it spare the collecting system? The Journal of urology 179(3),

1150-1154.

Lang, R., Patel, D., Morris, J.J., Rutschman, R.L., and Murray, P.J. (2002). Shaping

gene expression in activated and resting primary macrophages by IL-10. The

Journal of Immunology 169(5), 2253-2263.

Lapeyre-Prost, A., Terme, M., Pernot, S., Pointet, A.-L., Voron, T., Tartour, E., et al.

(2017). "Immunomodulatory activity of VEGF in cancer," in International review of

cell and molecular biology. Elsevier), 295-342.

Page 325: Determining the Oncological and Immunological Effects of

288

Leuchte, K., Staib, E., Thelen, M., Gödel, P., Lechner, A., Zentis, P., et al. (2020).

Microwave ablation enhances tumor-specific immune response in patients with

hepatocellular carcinoma. Cancer Immunology, Immunotherapy, 1-15.

Levings, M.K., Bacchetta, R., Schulz, U., and Roncarolo, M.G. (2002). The role of IL-10

and TGF-β in the differentiation and effector function of T regulatory cells.

International archives of allergy and immunology 129(4), 263-276.

Lin, K.W., Kim, Y., Maxwell, A.D., Wang, T.Y., Hall, T.L., Xu, Z., et al. (2014). Histotripsy

beyond the intrinsic cavitation threshold using very short ultrasound pulses:

microtripsy. IEEE Trans Ultrason Ferroelectr Freq Control 61(2), 251-265. doi:

10.1109/TUFFC.2014.6722611.

Liu, S., Zhang, C., Wang, B., Zhang, H., Qin, G., Li, C., et al. (2021). Regulatory T cells

promote glioma cell stemness through TGF-β-NF-κB-IL6-STAT3 signaling.

Cancer Immunol Immunother. doi: 10.1007/s00262-021-02872-0.

Lu, P., Zhu, X.-Q., Xu, Z.-L., Zhou, Q., Zhang, J., and Wu, F. (2009). Increased

infiltration of activated tumor-infiltrating lymphocytes after high intensity focused

ultrasound ablation of human breast cancer. Surgery 145(3), 286-293.

Mabuchi, S., Sasano, T., and Komura, N. (2021). Targeting Myeloid-Derived

Suppressor Cells in Ovarian Cancer. Cells 10(2). doi: 10.3390/cells10020329.

Mimura, K., Teh, J.L., Okayama, H., Shiraishi, K., Kua, L.F., Koh, V., et al. (2018). PD-

L1 expression is mainly regulated by interferon gamma associated with JAK-

STAT pathway in gastric cancer. Cancer Sci 109(1), 43-53. doi:

10.1111/cas.13424.

Page 326: Determining the Oncological and Immunological Effects of

289

Minami, Y., Nishida, N., and Kudo, M. (2019). Radiofrequency ablation of liver

metastasis: potential impact on immune checkpoint inhibitor therapy. European

radiology 29(9), 5045-5051.

Najafi, M., Hashemi Goradel, N., Farhood, B., Salehi, E., Nashtaei, M.S., Khanlarkhani,

N., et al. (2019). Macrophage polarity in cancer: A review. Journal of cellular

biochemistry 120(3), 2756-2765.

Ni, L., and Lu, J. (2018). Interferon gamma in cancer immunotherapy. Cancer medicine

7(9), 4509-4516.

Nobler, M.P. (1969). The abscopal effect in malignant lymphoma and its relationship to

lymphocyte circulation. Radiology 93(2), 410-412.

Ostuni, R., Kratochvill, F., Murray, P.J., and Natoli, G. (2015). Macrophages and cancer:

from mechanisms to therapeutic implications. Trends in immunology 36(4), 229-

239.

Pahk, K.J., Shin, C.-H., Bae, I.Y., Yang, Y., Kim, S.-H., Pahk, K., et al. (2019). Boiling

Histotripsy-induced Partial Mechanical Ablation Modulates Tumour

Microenvironment by Promoting Immunogenic Cell Death of Cancers. Scientific

reports 9(1), 1-12.

Pandit, H., Hong, Y.K., Li, Y., Rostas, J., Pulliam, Z., Li, S.P., et al. (2019). Evaluating

the regulatory immunomodulation effect of irreversible electroporation (IRE) in

pancreatic adenocarcinoma. Annals of surgical oncology 26(3), 800-806.

Pang, L., Han, S., Jiao, Y., Jiang, S., He, X., and Li, P. (2017). Bu Fei Decoction

attenuates the tumor associated macrophage stimulated proliferation, migration,

Page 327: Determining the Oncological and Immunological Effects of

290

invasion and immunosuppression of non-small cell lung cancer, partially via IL-10

and PD-L1 regulation. International Journal of Oncology 51(1), 25-38.

Pillai, K., Akhter, J., Chua, T.C., Shehata, M., Alzahrani, N., Al-Alem, I., et al. (2015).

Heat sink effect on tumor ablation characteristics as observed in monopolar

radiofrequency, bipolar radiofrequency, and microwave, using ex vivo calf liver

model. Medicine 94(9).

Pisetsky, D.S. (2011). Cell death in the pathogenesis of immune-mediated diseases: the

role of HMGB1 and DAMP-PAMP complexes. Swiss medical weekly 141,

w13256.

Plate, J. (2011). Clinical trials of vaccines for immunotherapy in pancreatic cancer.

Expert review of vaccines 10(6), 825-836.

Qu, S., Worlikar, T., Felsted, A.E., Ganguly, A., Beems, M.V., Hubbard, R., et al. (2020).

Non-thermal histotripsy tumor ablation promotes abscopal immune responses

that enhance cancer immunotherapy. Journal for ImmunoTherapy of Cancer

8(1).

Ringel-Scaia, V.M., Beitel-White, N., Lorenzo, M.F., Brock, R.M., Huie, K.E.,

Coutermarsh-Ott, S., et al. (2019). High-frequency irreversible electroporation is

an effective tumor ablation strategy that induces immunologic cell death and

promotes systemic anti-tumor immunity. EBioMedicine 44, 112-125. doi:

10.1016/j.ebiom.2019.05.036.

Rodriguez, P.C., Ernstoff, M.S., Hernandez, C., Atkins, M., Zabaleta, J., Sierra, R., et al.

(2009). Arginase I-producing myeloid-derived suppressor cells in renal cell

Page 328: Determining the Oncological and Immunological Effects of

291

carcinoma are a subpopulation of activated granulocytes. Cancer Res 69(4),

1553-1560. doi: 10.1158/0008-5472.Can-08-1921.

Saccomandi, P., Lapergola, A., Longo, F., Schena, E., and Quero, G. (2018). Thermal

ablation of pancreatic cancer: A systematic literature review of clinical practice

and pre-clinical studies. International Journal of Hyperthermia 35(1), 398-418.

Salama, P., Phillips, M., Grieu, F., Morris, M., Zeps, N., Joseph, D., et al. (2009).

Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance

in colorectal cancer. Journal of clinical oncology 27(2), 186-192.

Sanchez-Ortiz, R.F., Tannir, N., Ahrar, K., and Wood, C.G. (2003). Spontaneous

regression of pulmonary metastases from renal cell carcinoma after radio

frequency ablation of primary tumor: in situ tumor vaccine? The Journal of

urology 170(1), 178-179.

Schade, G.R. (2018). Evaluation of the Systemic Response to Boiling Histotripsy

Treatment for Renal Carcinoma.

Schade, G.R., Wang, Y.-N., D'Andrea, S., Hwang, J.H., Liles, W.C., and Khokhlova,

T.D. (2019). Boiling Histotripsy Ablation of Renal Cell Carcinoma in the Eker Rat

Promotes a Systemic Inflammatory Response. Ultrasound in medicine & biology

45(1), 137-147.

Scheffer, H.J., Stam, A.G., Geboers, B., Vroomen, L.G., Ruarus, A., de Bruijn, B., et al.

(2019). Irreversible electroporation of locally advanced pancreatic cancer

transiently alleviates immune suppression and creates a window for antitumor T

cell activation. Oncoimmunology 8(11), 1652532.

Page 329: Determining the Oncological and Immunological Effects of

292

Senn, N. (1903). Case of Splenomedullary Leukaemia Successfully Treated by the Use

of the Roentgen. Med Rec 64, 281-282.

Shadhu, K., and Xi, C. (2019). Inflammation and pancreatic cancer: An updated review.

Saudi J Gastroenterol 25(1), 3-13. doi: 10.4103/sjg.SJG_390_18.

Shao, Q., O'Flanagan, S., Lam, T., Roy, P., Pelaez, F., Burbach, B.J., et al. (2019).

Engineering T cell response to cancer antigens by choice of focal therapeutic

conditions. International Journal of Hyperthermia 36(1), 130-138.

Smolock AR, C.M., Vlaisavljevich E, Gendron-Fitzpatrick A, Green C, Ziemlewicz TJ

and Lee FT (2018). Robotically Assisted Sonic Therapy as a Non-Invasive Non-

Thermal Ablation Modality: Proof of Concept in a Porcine Liver Model. Radiology

287(2), 485-493.

Smolock, A.R., Cristescu, M.M., Vlaisavljevich, E., Gendron-Fitzpatrick, A., Green, C.,

Cannata, J., et al. (2018). Robotically Assisted Sonic Therapy as a Noninvasive

Nonthermal Ablation Modality: Proof of Concept in a Porcine Liver Model.

Radiology 287(2), 485-493. doi: 10.1148/radiol.2018171544.

Styn, N.R., Hall, T.L., Fowlkes, J.B., Cain, C.A., and Roberts, W.W. (2012). Histotripsy

of renal implanted VX-2 tumor in a rabbit model: investigation of metastases.

Urology 80(3), 724-729.

Summers, C., Rankin, S.M., Condliffe, A.M., Singh, N., Peters, A.M., and Chilvers, E.R.

(2010). Neutrophil kinetics in health and disease. Trends in immunology 31(8),

318-324.

Tanaka, A., and Sakaguchi, S. (2019). Targeting Treg cells in cancer immunotherapy.

European journal of immunology 49(8), 1140-1146.

Page 330: Determining the Oncological and Immunological Effects of

293

Tesi, R. (2019). MDSC; the most important cell you have never heard of. Trends in

pharmacological sciences 40(1), 4-7.

Thanos, L., Mylona, S., Galani, P., Pomoni, M., Pomoni, A., and Koskinas, I. (2008).

Overcoming the heat-sink phenomenon: successful radiofrequency thermal

ablation of liver tumors in contact with blood vessels. Diagnostic and

Interventional Radiology 14(1), 51.

Trellakis, S., Farjah, H., Bruderek, K., Dumitru, C., Hoffmann, T., Lang, S., et al. (2011).

Peripheral blood neutrophil granulocytes from patients with head and neck

squamous cell carcinoma functionally differ from their counterparts in healthy

donors. International journal of immunopathology and pharmacology 24(3), 683-

693.

van Beijnum, J.R., Buurman, W.A., and Griffioen, A.W. (2008). Convergence and

amplification of toll-like receptor (TLR) and receptor for advanced glycation end

products (RAGE) signaling pathways via high mobility group B1 (HMGB1).

Angiogenesis 11(1), 91-99. doi: 10.1007/s10456-008-9093-5.

van Raam, B.J., Drewniak, A., Groenewold, V., van den Berg, T.K., and Kuijpers, T.W.

(2008). Granulocyte colony-stimulating factor delays neutrophil apoptosis by

inhibition of calpains upstream of caspase-3. Blood, The Journal of the American

Society of Hematology 112(5), 2046-2054.

Vizio, B., Novarino, A., Giacobino, A., Cristiano, C., Prati, A., Ciuffreda, L., et al. (2012).

Potential plasticity of T regulatory cells in pancreatic carcinoma in relation to

disease progression and outcome. Experimental and therapeutic medicine 4(1),

70-78.

Page 331: Determining the Oncological and Immunological Effects of

294

Vlaisavljevich, E., Greve, J., Cheng, X., Ives, K., Shi, J., Jin, L., et al. (2016a). Non-

invasive ultrasound liver ablation using histotripsy: Chronic study in an in vivo

rodent model. Ultrasound in medicine & biology 42(8), 1890-1902.

Vlaisavljevich, E., Kim, Y., Allen, S., Owens, G., Pelletier, S., Cain, C., et al. (2013a).

Image-guided non-invasive ultrasound liver ablation using histotripsy: Feasibility

study in an in vivo porcine model. Ultrasound in medicine & biology 39(8), 1398-

1409.

Vlaisavljevich, E., Kim, Y., Owens, G., Roberts, W., Cain, C., and Xu, Z. (2013b).

Effects of tissue mechanical properties on susceptibility to histotripsy-induced

tissue damage. Physics in Medicine & Biology 59(2), 253.

Vlaisavljevich, E., Kim, Y., Owens, G., Roberts, W., Cain, C., and Xu, Z. (2014a).

Effects of tissue mechanical properties on susceptibility to histotripsy-induced

tissue damage. Physics in Medicine and Biology 59(2), 253-270. doi:

10.1088/0031-9155/59/2/253.

Vlaisavljevich, E., Lin, K.W., Maxwell, A., Warnez, M., Mancia, L., Singh, R., et al.

(2015a). Effects of Ultrasound Frequency and Tissue Stiffness on the Histotripsy

Intrinsic Threshold for Cavitation. Ultrasound Med Biol.

Vlaisavljevich, E., Maxwell, A., Mancia, L., Johnsen, E., Cain, C., and Xu, Z. (2016b).

Visualizing the Histotripsy Process: Bubble Cloud-Cancer Cell Interactions in a

Tissue-Mimicking Environment. Ultrasound Med Biol. doi: S0301-5629(16)30099-

0 [pii] 10.1016/j.ultrasmedbio.2016.05.018.

Vlaisavljevich, E., Maxwell, A., Warnez, M., Johnsen, E., Cain, C., and Xu, Z. (2014b).

Histotripsy-induced cavitation cloud initiation thresholds in tissues of different

Page 332: Determining the Oncological and Immunological Effects of

295

mechanical properties. IEEE Transactions on Ultrasonics, Ferroelectrics, and

Frequency Control 61(2), 341-352. doi: 10.1109/TUFFC.2014.6722618.

Vlaisavljevich, E., Owens, G., Lundt, J., Teofilovic, D., Ives, K., Duryea, A., et al. (2017).

Non-invasive liver ablation using histotripsy: preclinical safety study in an in vivo

porcine model. Ultrasound in medicine & biology 43(6), 1237-1251.

Vlaisavljevich, E., Xu, Z., Arvidson, A., Jin, L., Roberts, W., and Cain, C. (2015b).

Effects of thermal preconditioning on tissue susceptibility to histotripsy.

Ultrasound in medicine & biology 41(11), 2938-2954.

Wan, S., Zhao, E., Kryczek, I., Vatan, L., Sadovskaya, A., Ludema, G., et al. (2014).

Tumor-associated macrophages produce interleukin 6 and signal via STAT3 to

promote expansion of human hepatocellular carcinoma stem cells.

Gastroenterology 147(6), 1393-1404.

Wang, D., An, G., Xie, S., Yao, Y., and Feng, G. (2016). The clinical and prognostic

significance of CD14+ HLA-DR−/low myeloid-derived suppressor cells in

hepatocellular carcinoma patients receiving radiotherapy. Tumor Biology 37(8),

10427-10433.

Wang, D., Zhang, X., Gao, Y., Cui, X., Yang, Y., Mao, W., et al. (2020). Research

Progress and Existing Problems for Abscopal Effect. Cancer Manag Res 12,

6695-6706. doi: 10.2147/cmar.S245426.

Wang, X., and Lin, Y. (2008). Tumor necrosis factor and cancer, buddies or foes? 1.

Acta Pharmacologica Sinica 29(11), 1275-1288.

Page 333: Determining the Oncological and Immunological Effects of

296

Watford, W.T., Moriguchi, M., Morinobu, A., and O'Shea, J.J. (2003). The biology of IL-

12: coordinating innate and adaptive immune responses. Cytokine Growth Factor

Rev 14(5), 361-368. doi: 10.1016/s1359-6101(03)00043-1.

Widenmeyer, M., Shebzukhov, Y., Haen, S.P., Schmidt, D., Clasen, S., Boss, A., et al.

(2011). Analysis of tumor antigen‐specific T cells and antibodies in cancer

patients treated with radiofrequency ablation. International Journal of Cancer

128(11), 2653-2662.

Wilky, B.A. (2019). Immune checkpoint inhibitors: the linchpins of modern

immunotherapy. Immunological reviews 290(1), 6-23.

Wislez, M., Fleury-Feith, J., Rabbe, N., Moreau, J., Cesari, D., Milleron, B., et al. (2001).

Tumor-derived granulocyte-macrophage colony-stimulating factor and

granulocyte colony-stimulating factor prolong the survival of neutrophils

infiltrating bronchoalveolar subtype pulmonary adenocarcinoma. The American

journal of pathology 159(4), 1423-1433.

Wissniowski, T.T., Hänsler, J., Neureiter, D., Frieser, M., Schaber, S., Esslinger, B., et

al. (2003). Activation of tumor-specific T lymphocytes by radio-frequency ablation

of the VX2 hepatoma in rabbits. Cancer research 63(19), 6496-6500.

Worlikar, T., Mendiratta-Lala, M., Greve, J., Cho, C., Hubbard, R., Vlaisavljevich, E., et

al. (2019). "Non-Invasive Orthotopic Liver Tumor Ablation Using Histotripsy in an

in Vivo Rodent Hepatocellular Carcinoma (HCC) Model", in: Society of

Interventional Oncology. (Boston, MA, USA).

Worlikar, T., Vlaisavljevich, E., Gerhardson, T., Greve, J., Wan, S., Kuruvilla, S., et al.

(2018). Histotripsy for Non-Invasive Ablation of Hepatocellular Carcinoma (HCC)

Page 334: Determining the Oncological and Immunological Effects of

297

Tumor in a Subcutaneous Xenograft Murine Model. Conf Proc IEEE Eng Med

Biol Soc 2018, 6064-6067. doi: 10.1109/EMBC.2018.8513650.

Wu, F., Wang, Z.-B., Lu, P., Xu, Z.-L., Chen, W.-Z., Zhu, H., et al. (2004). Activated anti-

tumor immunity in cancer patients after high intensity focused ultrasound

ablation. Ultrasound in medicine & biology 30(9), 1217-1222.

Wu, K., Lin, K., Li, X., Yuan, X., Xu, P., Ni, P., et al. (2020). Redefining Tumor-

Associated Macrophage Subpopulations and Functions in the Tumor

Microenvironment. Front Immunol 11, 1731. doi: 10.3389/fimmu.2020.01731.

Wu, L., Saxena, S., Awaji, M., and Singh, R.K. (2019). Tumor-associated neutrophils in

cancer: going pro. Cancers 11(4), 564.

Wu, L., and Zhang, X.H. (2020). Tumor-Associated Neutrophils and Macrophages-

Heterogenous but Not Chaotic. Front Immunol 11, 553967. doi:

10.3389/fimmu.2020.553967.

Xie, C., Duffy, A.G., Mabry‐Hrones, D., Wood, B., Levy, E., Krishnasamy, V., et al.

(2019). Tremelimumab in combination with microwave ablation in patients with

refractory biliary tract cancer. Hepatology 69(5), 2048-2060.

Xing, Y., Lu, X., Pua, E.C., and Zhong, P. (2008). The effect of high intensity focused

ultrasound treatment on metastases in a murine melanoma model. Biochemical

and biophysical research communications 375(4), 645-650.

Yang, R., Reilly, C.R., Rescorla, F.J., Sanghvi, N.T., Fry, F.J., Franklin Jr, T.D., et al.

(1992). Effects of high-intensity focused ultrasound in the treatment of

experimental neuroblastoma. Journal of pediatric surgery 27(2), 246-251.

Page 335: Determining the Oncological and Immunological Effects of

298

Yin, J., Dong, J., Gao, W., and Wang, Y. (2018). A case report of remarkable response

to association of radiofrequency ablation with subsequent Atezolizumab in stage

IV nonsmall cell lung cancer. Medicine 97(44).

Youn, J.I., and Gabrilovich, D.I. (2010). The biology of myeloid‐derived suppressor cells:

the blessing and the curse of morphological and functional heterogeneity.

European journal of immunology 40(11), 2969-2975.

Zamarron, B.F., and Chen, W. (2011). Dual roles of immune cells and their factors in

cancer development and progression. International journal of biological sciences

7(5), 651.

Zhou, H., Jiang, M., Yuan, H., Ni, W., and Tai, G. (2021). Dual roles of myeloid-derived

suppressor cells induced by Toll-like receptor signaling in cancer. Oncol Lett

21(2), 149. doi: 10.3892/ol.2020.12410.

Zhou, K., Cheng, T., Zhan, J., Peng, X., Zhang, Y., Wen, J., et al. (2020). Targeting

tumor-associated macrophages in the tumor microenvironment. Oncol Lett 20(5),

234. doi: 10.3892/ol.2020.12097.

Page 336: Determining the Oncological and Immunological Effects of

299

Chapter 8. Histotripsy Ablation Alters the Tumor Microenvironment and Promotes Immune System Activation in a Subcutaneous Model of Pancreatic

Cancer

Alissa Hendricks-Wenger, Jaqueline Sereno, Jessica Gannon, Allison Zeher, Rebecca M Brock, Natalie Betiel-White, Alexander Simon, Rafael V. Davalos, Sheryl

Coutermarsh-Ott, Eli Vlaisavljevich, Irving C. Allen

This chapter is excerpted from a manuscript published in IEEE Transactions in UFFC.

2021 IEEE. Reprinted, with permission, from: A. Hendricks-Wenger et al., "Histotripsy Ablation Alters the Tumor Microenvironment and Promotes Immune System Activation in a Subcutaneous Model of Pancreatic Cancer," in IEEE Transactions on Ultrasonics,

Ferroelectrics, and Frequency Control.

Page 337: Determining the Oncological and Immunological Effects of

300

8.1 Abstract

Pancreatic cancer is a significant cause of cancer-related deaths in the United States

with an abysmal 5-year overall survival rate that is under 9%. Reasons for this mortality

include the lack of late-stage treatment options and the immunosuppressive tumor

microenvironment. Histotripsy is an ultrasound-guided, non-invasive, non-thermal tumor

ablation therapy that mechanically lyses targeted cells. To study the effects of

histotripsy on pancreatic cancer, we utilized an in vitro model of pancreatic

adenocarcinoma and compared the release of potential antigens following histotripsy

treatment to other ablation modalities. Histotripsy was found to release immune-

stimulating molecules at magnitudes similar to other non-thermal ablation modalities

and superior to thermal ablation modalities, which corresponded to increased innate

immune system activation in vivo. In subsequent in vivo studies, murine Pan02 tumors

were grown in mice and treated with histotripsy. Flow cytometry and rtPCR were used

to determine changes in the tumor microenvironment over time compared to untreated

animals. In mice with pancreatic tumors, we observed significantly increased tumor-

progression-free and general survival, with increased activation of the innate immune

system 24 hours post-treatment and decreased tumor-associated immune cell

populations within 14 days of treatment. This study demonstrates the feasibility of using

histotripsy for pancreatic cancer ablation and provides mechanistic insight into the initial

innate immune system activation following treatment. Further work is needed to

Page 338: Determining the Oncological and Immunological Effects of

301

establish the mechanisms behind the immunomodulation of the tumor

microenvironment and immune effects.

8.2 Keywords

Biological Effects & Dosimetry, Therapeutics

Page 339: Determining the Oncological and Immunological Effects of

302

8.3 Introduction

Pancreatic cancer is the fourth leading cause of cancer-related deaths, with a 9%

survival rate due to its late diagnosis and lack of curative treatment options [1, 2].

Standard treatment options are limited and include surgery, chemotherapy, and

radiation [3]. Only 20% of patients have tumors that can be surgically removed, and of

those patients the cure rate is less than 25% [4]. Ablation procedures can act as a

replacement for, or as an adjuvant to, surgery. The most commonly used ablation

modalities for treating cancers in abdominal organs are radiofrequency ablation (RFA),

microwave ablation, high-intensity focused ultrasound, and cryoablation. RFA is a

thermal, minimally-invasive ablation modality that utilizes high-frequency alternating

currents to thermally induce thermal necrosis [5]. Microwave ablation, also thermal and

minimally-invasive, induces cell death through electromagnetic microwaves that

produce friction and heat [6]. High-intensity focused ultrasound ablates cells by

depositing ultrasound energy at a focal point to rapidly increase tissue temperature [7].

Cryoablation results in tumor cell destruction through ice-crystal formation as a result of

liquid nitrogen or argon gas delivered to the tissue and does not result in protein

denaturation, which has led to increased reports of immunological effects compared to

RFA and microwave ablation [5]. The thermal ablation modalities, although efficacious

in treating certain malignancies, have not yet been ubiquitously accepted into clinics for

pancreatic cancer therapy due to the risk of thermal damage to healthy pancreatic

tissue, vasculature, and other critical structures.

Page 340: Determining the Oncological and Immunological Effects of

303

Recent advancements have established non-thermal therapies that have the

potential to treat pancreatic cancer. For example, irreversible electroporation (IRE)

utilizes short, high-voltage electrical pulses that non-thermally open micro-pores in cell

plasma membranes, inducing cell death [8]. Clinical trials have shown that IRE can

ablate pancreatic tumors without damaging nearby critical structures and have had

dramatic effects on patient survival that may be due to the induction of

immunomodulatory mechanisms [9-14]. Given IRE’s controlled cell death mechanisms,

the procedure has been found to release immunostimulatory molecules and lead to a

more immunologically active tumor microenvironment after treatment [15]. Furthermore,

recent studies comparing the thermal ablation modalities, cryoablation, and IRE have

found that the non-thermal modalities are more potent at stimulating an anti-tumor

microenvironment [16]. These procedures still involve surgical incisions that increase

the possibility of surgery-related injury or infection.

To address the clinical limitations of IRE and other ablation modalities, new

focused ultrasound ablation methods have been studied as a completely non-invasive,

non-thermal alternative. Histotripsy is a non-thermal, non-ionizing, image-guided

ablation modality that uses focused-ultrasound to initiate acoustic cavitation, which

leads to the lysis of cells contained in the targeted area [17, 18]. Ablation of internal

targets with histotripsy is effective with little to no off-target effects [19-21]. Early studies

with histotripsy ablation of melanoma, hepatocellular carcinoma, renal cell carcinomas,

colorectal carcinomas, and neuroblastomas established that there is an activation of

local, cellular, and systemic immune responses [22-27].

Page 341: Determining the Oncological and Immunological Effects of

304

In this study, we assess the ability of histotripsy to ablate subcutaneous

pancreatic tumors and determine the immunological changes within the treated tumors

over time. First, we compared the release of damage-associated molecular patterns

(DAMPs) known to activate the innate immune system and neoantigen generation

following histotripsy against other tumor ablation modalities. We extended these in vitro

findings using the in vivo Pan02 murine pancreatic cancer model. This mouse model

was chosen given its well characterized progression and described immune effects to

cancer therapies [15, 28, 29]. Using this model, we demonstrate effective tumor

ablation, pro-inflammatory changes in the tumor micro-environment, and identify critical

immune signaling mechanisms associated with histotripsy treatment.

8.4 Procedures

8.4.1 Tumor Injections and Monitoring

All in vivo experiments were conducted under institutional IACUC approval and

following the NIH Guide for the Care and Use of Laboratory Animals. For these studies,

male and female mice were equally utilized in the 7 to 10-week age range. Once the

entire cohort of C57/Bl6 mice reached a minimum weight of 20g, 100 µL of Pan02 cells

(DTP, DCTD Tumor Repository) at a concentration of 6.0x107 cells/mL of Matrigel

(Corning) were injected into the right flank of the mice that were anesthetized with

vaporized isoflurane (1.5 L/min oxygen flow with 1-3% isoflurane). Control animals were

injected with the same amount of Matrigel that did not contain Pan02 cells. The mice

Page 342: Determining the Oncological and Immunological Effects of

305

were then monitored three times a week until the end of the study. Tumor diameters

were measured with calipers and calculated as the square root of two perpendicular

measurements, as previously described [24]. The weights and tumor sizes were

recorded along with the general health of the mice.

8.4.2 Histotripsy Set Up

In vivo studies used a custom 1MHz, 8-element small animal histotripsy transducer

with a geometric focus of 36 mm, an aperture size of 52.7 mm, and an f-number of 0.68.

The full-width half-maximum (FWHM) dimensions at a geometric focus of this

transducer were 0.98 mm, 0.93 mm, and 3.9 mm in transverse, elevational, and axial,

respectively. The transducer was driven via a custom high-voltage pulser designed to

generate short therapy pulses of <2 cycles controlled by a field-programmable gate

array (FPGA) board (Altera DE0-Nano Terasic Technology, Dover, DE, USA)

programmed for histotripsy therapy pulsing. The transducer was positioned in a tank of

degassed water heated to 37+4ºC beneath a custom-designed mouse surgical stage

(Fig.1A) and attached to a computer-guided 3-D positioning system with a 0.05 mm

motor resolution to control the automated volumetric treatments. A linear ultrasound

imaging probe with a frequency range of 10-18 MHz (L18-10L30H-4, Telemed,

Lithuania, EU) was coaxially aligned inside the transducer for treatment guidance and

monitoring [20, 24]. The transducer was powered by a high voltage DC power supply

(GENH750W, TDK-Lambda), and the system was controlled using a custom user-

interface operated through MATLAB (MathWorks).

Page 343: Determining the Oncological and Immunological Effects of

306

Figure 8-1 In Vivo Experimental Setup. Murine histotripsy experiments were conducted using a 1Mhz transducer (A). Timing for treatment, euthanasia, and data collection were set as diagramed (B). Days with histology collection are noted by cassettes, flash frozen tumors noted by tubes with tumors, serum noted by tubes with serum, and flow cytometry is noted by flow charts. Tumors located with ultrasound imaging were used for planning automated ablation disks (C) and raster scan plots (D). Therapy was guided by co-axially aligned ultrasound imaging (E). Red arrow indicates a bubble cloud that was generated during treatment.

Page 344: Determining the Oncological and Immunological Effects of

307

8.4.3 n Vitro Ablations Treatment Parameters

Pan02 cells transfected with a plasmid that produces the influenza antigen

hemagglutinin (Pan02-HA) were used for these in vitro experiments. HA is a common

surrogate used in tumor-specific antigen studies. Pan02-HA cells were ablated in four

treatments and two levels of ablation (Fig.2A). Cells were collected, centrifuged to

pellet, and were resuspended in PBS at a concentration of 10x106 cells/mL. 1 mL of

cell suspension was used for each treatment. All samples were kept on ice until

treatment. For all treatments, the partial ablation dosages were determined to be a

successful ablation with >30% viability and the full ablation dosages had <10% viability

for all samples. For each ablation modalities the prefix “f-” is added before the modality

name within the text to refer to the full ablation, and “p-” is added to refer to the partial

ablation. For example, f-histotripsy is full ablation histotripsy and p-Histotripsy is partial

ablation histotripsy.

Histotripsy treatments were done in a custom holder (Fig.2B), utilizing the same

1MHz transducer from the in vivo work at a PRF of 250Hz at a single spot for 0.5 or 5

minutes. The movement of the transducer during treatment was not necessary given the

circulation of the cell suspension caused by histotripsy that was observed during

treatment and in previous studies [30]. For cryoablation, cells were placed in liquid

nitrogen (approximately -160oC) for 30 minutes. No lower dose was used given that any

drop to therapeutically low temperatures (-20oC to -190oC) resulted in high levels of cell

death [31]. Thermal ablation consisted of cells being kept at 80oC on a heating block for

1 or 30 minutes. The protocols for cryoablation and thermal ablation were based upon a

Page 345: Determining the Oncological and Immunological Effects of

308

previous study.[16] For samples not included in analysis, the temperatures of the

thermally treated samples were confirmed with a thermometer to reach 45oC, minimum

temperature to be considered thermal ablation, within the first half a minute and 80oC by

3 minutes [32]. Samples frozen with liquid nitrogen were assumed to surpass the

therapeutic temperature threshold. Given the extensive prior studies showing no

temperature change with IRE at the prescribed dosages or histotripsy no temperature

measurements were done for these therapies [17, 33-35]. For experimental sham,

untreated control samples of cells were prepared and handled identically to treated

samples, but instead of receiving treatment remained on wet ice during the treatment of

other cells.

Figure 8-2 Treatment flow and dosages for various ablation modalities (A). Histotripsy experiments on cell suspensions were conducted using a 1Mhz transducer (B).

Page 346: Determining the Oncological and Immunological Effects of

309

For IRE treatments, cells were suspended in a sucrose solution, described

previously [36] to improve the quality of electrical transduction was adjusted to use a

base of PBS to mitigate differences in treatment groups, and placed into 4 mm cuvettes.

A generator (BTX ECM 830, Harvard Apparatus, Holliston, MA) was used to apply 100

pulses with widths of 100 s at a frequency of 1 Hz and an electric field of either 500

V/cm or 2000 V/cm. Cells were then cultured for 24 hours before supernatant collection

to allow for the controlled cell death of IRE to take place [15]. Additional controls for IRE

samples to accommodate for the extra downstream processing. In these cases, the

media that was used to culture the IRE samples over night was also ran through BCA

and nanodrop. The resulting values from the controls were subtracted from the IRE

samples to accommodate for the excess signaling caused by the media in culturing.

Viability for all modalities was determined shortly after treatment with standard Trypan

Blue counting with a 1:40 dilution due to the high concentration of cells, and calculated

as a percentage of cells remaining viable after treatment. Post-ablation samples were

centrifuged at 1000xg for 5 minutes and supernatants were collected. Supernatants for

cryoablation, thermal ablation, and histotripsy were collected immediately after

treatment and IRE 24 hours after incubation at 37oC.

8.4.4 In Vivo Histotripsy Treatment

Table 8-1 Subject Numbers Per Experimental Group

Immediate Group

Acute Group Chronic Group

Flow Cytometry

No Treatment No Tumor - 3 3 -

Tumor 3 7 7 4/time point

Histotripsy Treated Tumors 3 7 7 4/time point

Page 347: Determining the Oncological and Immunological Effects of

310

Mice were treated when the average tumor diameter of the cohort was

approximately 0.6cm, to ensure that the smallest tumors were large enough for

targeting (Fig.1B). Animals were euthanized 24 hours after treatment, referred to as the

acute group, (n=7 treated, n=7 untreated controls, and n=3 tumor-free untreated

controls) or at survival time points, referred to as the chronic group (n=7 treated, n=7

untreated controls, and n=3 tumor-free untreated controls) (Table 1). Euthanasia of

chronic group animals, the point determined as their general survival, was determined

as either 1) when clinical health evaluations found deleterious symptoms including

hunching, irregular respiratory rate and rhythm, decreased alertness and socialization,

or poor extremity utilization or 2) when the tumor diameter exceeded 1.4cm. At

necropsy, the immediate group (n=3) mice that were treated and untreated (Table 1)

were euthanized and tumors were formalin fixed for histopathology to determine efficacy

of treatment. For the acute and chronic group mice, serum was collected and the

tumors were sectioned with a portion formalin fixed for histopathology and another

portion flash frozen for mRNA analysis (Fig.1B).

Prior to each treatment, each mouse had fur removed over the tumors using the

depilatory cream Nair (Naircare, Ewing, NJ). Mice were anesthetized with vaporized 2-

4% isoflurane with an oxygen flow rate of 1.5 L/min. The mice were then placed on the

subject stage with their tumor submerged in the degassed water in the subject stage’s

window. The tumor was located using the ultrasound imaging probe that was coaxially

aligned to the histotripsy transducer, and then targeted using an automated volumetric

ablation algorithm which controlled the treatment following manual targeting.

Page 348: Determining the Oncological and Immunological Effects of

311

For each tumor, a 3D ellipsoidal volume was targeted using conservative margins of

approximately 0.5-2mm from the skin and underlying tissues (muscle, intestines, etc).

Since our automated treatment is a perfect ellipsoid and tumors are not, there was

some small amount of variation between subjects. Using these margins, we intentionally

targeted a partial ablation of approximately 60-75% of the tumor volume. This volume

consisted of multiple, concentric 2D ellipical slices. Each treatment slice was separated

0.75mm apart. Within each slice, the area was populated with grid points separated

0.75mm apart in the transverse direction and 1.25 mm apart in the axial direction

(Fig.1C). At each treatment point, histotripsy was applied at a pulse repetition frequency

(PRF) of 250Hz and a dwell time of 1 second, consequently sending 250 pulses to each

point. For each slice, the automated treatment first generated a histotripsy bubble cloud

at the center point of the slice and then scanned in a raster pattern to cover one half of

the slice area. The transducer was then returned to the center of the ellipse and

scanned in a raster pattern to cover the other half of the slice. Once a slice was

scanned through completely, the system proceeds to the next slice, which is repeated

until the entire ellipsoidal volume was treated. Throughout treatment, ultrasound

guidance confirmed the location of the bubble cloud for the duration of the volumetric

ablation (Fig.1D). After treatment, the tumor volume was again imaged with ultrasound

imaging in order to assess for tissue ablation.

Page 349: Determining the Oncological and Immunological Effects of

312

8.4.5 Determining DNA Release and Quality

Each collected sample for each treatment group (n=5 for the no treatment group,

n=9-11/treatment group) was analyzed with a nanodrop drop, and the DNA

concentrations (ng/μL) and the 260/280 absorbance ratios were recorded. Samples

were then run on an ethidium bromide gel to visualize DNA strand sizes present in the

samples utilizing a 100bp HyperLadder (Bioline) and following standard protocol.

8.4.6 Determining Protein Release and Quality

The protein released was quantified using a BCA assay (Thermo Scientific) following

the manufacturer's protocol. Western blots were run using pre-made gels (Thermo

Scientific) following standard protocols with 5ug of protein/sample and transferred with

the iBlot 2 Dry Blotting System (Thermo Scientific). HA antigen release was determined

using an HA antibody (Cell Signaling) as per the manufacturer’s protocols. The protein’s

band area was quantified using iBright Analysis Software (n=5/treatment group).

8.4.7 Histopathology

Tissues were harvested from animals and fixed in 10% formalin. Paraffin-embedded

formalin-fixed tissues were stained with hematoxylin and eosin (H&E) following standard

protocols. Evaluations were performed by trained individuals and independently verified

by a blinded, board-certified veterinary pathologist (S.C.O.).

Page 350: Determining the Oncological and Immunological Effects of

313

8.4.8 Profiling Gene Expression and Pathway Analysis

Using tumors flash-frozen from in vivo experiments, total RNA isolated from tumor

samples using the RNeasy Mini Kit (250) (Qiagen), where the manufacturer’s standard

protocol was followed. RNA levels were quantified with a nanodrop, converted to cDNA,

and were then pooled (n=7/tumor group and n=3/control group). Pooled cDNA was

placed into the commercially available pathway-focused array “Cancer Inflammation

and Immunity Crosstalk” (SuperArrayTM platform; Qiagen) following the manufacture’s

protocol (1/pooled groups). Fold change was determined using standard ΔΔCT

calculations. Gene expression was analyzed using IPA (integrated pathway analysis,

Qiagen) software to model changes in complex pathways [36].

Table 8-2 Flow Cytometry Immune Cell Markers

IMMUNE CELL TYPE IDENTIFYING MARKERS

Macrophages (MO) CD45+ CD11c+ F4/80+ Ly6C-

Inflammatory Dendritic Cells (iDCs) CD45+ CD11c+ F4/80- Ly6C+

Classic Dendritic Cells (cDCs) CD45+ CD11c+ F4/80- Ly6C-

Monocytic Myeloid Derived Suppressor Cells (M-MDSC) CD45+ CD11c- Ly6C+ Ly6G-

Granulocytes CD45+ CD11c- Ly6C- Ly6G+

CD8+ T cells CD45+ CD3+ CD4- CD8+

CD4+ T cells CD45+ CD3+ CD4+ CD8-

T Regulatory Cells (Treg cells) CD45+ CD3+ CD4+ CD8- FoxP3+

T Helper Cells (Th cells) CD45+ CD3+ CD4+ CD8- FoxP3-

Page 351: Determining the Oncological and Immunological Effects of

314

8.4.9 Flow Cytometry Panel Staining

For immune cell profiling, additional mice were injected with tumors and taken down

1, 7, and 14 days after treatment (Fig.1B) with an n=4 per treated and untreated control

group at each time point (Table 1).

After removal of the tumor, avoiding skin and fur, the tumor was placed into 8mL of

cold RPMI. Even though Pan02 tumors have microvasculature,[37] because they are

not very bloody due to total blood removal via heart stick, we did not perfuse the tissue

before harvest or processing. The tissue was then mechanically digested and strained

into a 50mL conical tube. After centrifuging at 300xg for 10 minutes at 4℃, the

supernatant was discarded and the pellet resuspended in 10mL of RPMI and was

plated onto a 96-well V-bottom plate at a density of 1.0x106 cells/well. A 1:200 dilution

of FACS buffer, sterile PBS with 2% FBS and 0.1% sodium azide, and anti-CD16/32

was added to the plate at a concentration of 50µL /well. Antibodies were diluted with

FACS buffer and added directly to the wells. The following antibodies were used: anti-

CD8 SuperBright 645, anti-CD45 SuperBright 645, anti-CD11c APC, anti-CD45 PE,

anti-F4/80 FITC, anti-CD4 PE.Cy5, anti-CD8 A488, anti-CD3 APC, anti-Ly6C APC Cy5,

anti-Ly6G PE, and anti-FOXP3 PerCP Cy. Cells were washed with PBS and evaluated

with FACS (BD Biosciences). Gating for specific immune cell populations is listed in

Table 2. It should be noted that the population ‘granulocytes’ is likely to contain both

granulocytic myeloid derived suppresser cells and neutrophils [38, 39]. Additionally, by

gating macrophages as both CD11c and F4/80 we are most likely focusing on a

Page 352: Determining the Oncological and Immunological Effects of

315

subpopulation of macrophages, more extensive panels would be necessary to confirm

the ratio of subpopulations.

8.4.10 HMGB1 Serum ELISA

Serum levels of high mobility group protein box 1 (HMGB1) in acute and survival

group mice were determined utilizing an ELISA assay kit (ABclonal) following the

manufactures suggested protocol.

8.4.11 Statistics

Data were analyzed using GraphPad Prism, version 8. Statistical significance was

defined as p ≤ 0.05, where values were not significant but p<0.20 the value is noted in

the text. All data are represented as the mean ± SEM. A Student's two-tailed t-test was

used when comparing two experimental groups. When many t-tests were performed for

one graph, group letter designations were used. Lowercase letters on top of bars

indicate significance; bars with the sample letter designation are not significant while

those that do not share a letter are significant (p<0.05). Multiple comparisons were done

using one-way or two-way ANOVA where appropriate, and then followed by Tukey post-

test for multiple pairwise examinations.

Page 353: Determining the Oncological and Immunological Effects of

316

8.5 Results

8.5.1 Different Ablation Modalities Show Differential Release of Damage Associated

Molecular Patterns (DAMPs) and Potential Antigens

Figure 8-3 Cell suspension ablations resulted in partial and full ablations (A). DNA release was quantified (B) and visualized (C). Protein release was quantified (D) and the relative release of the antigen HA was quantified from western blot bands (E). Lowercase letters on top of bars indicate significance; bars with the sample letter designation are not significant while those that do not share a letter are significant (p<0.05).

Pan02-HA cells were treated with thermal ablation, cryoablation, IRE, and histotripsy

at full (f-) and partial (p-) doses, and were chosen based on previous literature (Fig.2A)

[16]. Treatments that lead to less than 10% viability in all samples were considered fully

ablated and those that were greater than 30% in all samples were considered partially

Page 354: Determining the Oncological and Immunological Effects of

317

ablated (Fig.3A). All samples’ lysates were analyzed for peptide and DNA nucleotide

release (n=5 for no treatment, n=9-11 for treatment groups).

Extracellular DNA is a DAMP and often correlates with extracellular nuclear proteins,

such as HMGB1, that also acts as robust damage signaling. Here, we observed

increased levels of DNA for all treatments compared to untreated samples (Fig.3B).

DNA release was evaluated by nucleotide quantification, which showed f-IRE (470.73+/-

283.34 ng/ml) to have a significantly (p<0.05) higher concentration than most other

modalities, excluding f-cryoablation (262.49+/-159.00 ng/ml) and f-histotripsy (315.71+/-

175.28 ng/ml). P-thermal (177.33+/-113.15 ng/ml), f-thermal (170.48+/-72.20 ng/ml), p-

IRE (179.82+/-141.98 ng/ml), and p-histotripsy (165.03+83.09 ng/ml) were all similar to

each other. Only f-IRE and f-histotripsy were significantly greater than the untreated

control samples (20.35+/-10.11 ng/ml). Gel electrophoresis showed that cryoablation

and both dosages of histotripsy left large segments of detectable DNA, while untreated,

IRE and thermally ablated samples produced no visible bands (Fig.3C).

Peptide release, which correlates with potential antigens, was observed to be

released significantly in non-thermal ablation modalities (Fig.3D). There was no

significant difference between p-thermal (383.8+/-345.4 ug/ml) or f-thermal (443.3+/-

255.8 ug/ml) ablation’s peptide release from samples that were not treated (194.5+/-

50.7 ug/ml). On the other hand, f-IRE (2113+/-409.1 ug/ml) and f-histotripsy (2208+/-

751.8 ug/ml) were significantly (p<0.05) higher levels of release compared to all other

modalities, except for p-IRE (1869+/-431.3 ug/ml). F-cryoablation (1512+/-415.4 ug/ml)

and p-histotripsy (1300+/-490.0 ug/ml) were significantly different from the low

(significance group “a”) and high release clusters (significance group “c”).

Page 355: Determining the Oncological and Immunological Effects of

318

Release of HA was confirmed on western blot for all treated samples, regardless of

ablation treatment (Sup Fig.1). The average area of the various treatments HA bands

was not significantly different between any therapies compared to each other nor the

untreated samples (297.6+/-39.3 units) (Fig.3E). The highest detection of HA was found

in p-thermal treated samples (367.8+/-24.25 units), followed by p-histotripsy (344.3+/-

38.5 units) and f-histotripsy (340.0+/-35.2 units). F-cryoablation (332.0+/-56.4 units), f-

thermal (320.3+/-33.1 units), p-IRE (322.0+/-26.4 units), and f-IRE (317.2+/-24.2 units)

were all found to have larger bands than no treatment and smaller than p-thermal, p-

histotripsy, and f-histotripsy.

8.5.2 Histotripsy is an Effective Tumor Ablation Modality in the Subcutaneous Pan02

Model

The schematic shows the custom histotripsy rig and automated ablation procedure

that was used for in vivo treatments (Fig.1). Co-axial ultrasound imaging confirmed the

presence of a bubble cloud within the tumors during treatment (Fig.1E). Mice were

treated when tumors reached 0.6cm in diameter on average and harvested in groups as

depicted (Fig.1B). Ablation of tumors was confirmed with ultrasound, with increased

hypoechoic regions, and histopathology, where a partial ablation of tumors with viable

tumor tissue on margins and in islands within the ablation zone was observed (Fig.4).

On ultrasound images, the center of the treated region had a more notable decrease in

ultrasound reflection, while the dermal margin maintained a comparable hyperechocity

after treatment compared to before (Fig.4A-B). This pattern was also noted on

Page 356: Determining the Oncological and Immunological Effects of

319

histopathology. The center of untreated tumors has a characteristic necrotic core

(Fig.4C), and the treated tumors appeared to have a larger region of cell death that

extends nearly to the margins of the tumor (Fig.4D).

Figure 8-4 Ultrasound images of tumor before (A) and after (B) treatment, exhibiting more hypoechoic central region post-histotripsy. Orange shapes on ultrasound indicate location of tumors. Green circles indicate location of bubble cloud determined in water prior to treatment, and were utilized for treatment planning. Histology to tumors without (C) and with (D) treatment show decreased cellular detail after treatment. Dotted black line outlines the necrotic core of the tissue. Scale bar on H&E images = 500 um.

Page 357: Determining the Oncological and Immunological Effects of

320

Figure 8-5 After treatment of tumors on day 8, as indicated by the black arrow, the reduction of tumor volume was indicated by caliper measurements (A). Changes health observed in tumor-progression free survival (B) and general survival (C).

Calculated tumor diameters decreased for a few days after the partial ablation with

histotripsy and maintained size for two weeks before resuming tumor growth (Fig.5A).

The average tumor size of untreated tumors continued to increase in size at a relatively

steady rate (Fig.5A). The greatest difference between the treated and untreated groups

was reached on day 15, 8 days post-treatment when the treated tumors were 43% of

the size of the untreated tumors on average. Compared to the untreated mice,

histotripsy treatment increased tumor-progression-free survival by 19 days from 10 days

Page 358: Determining the Oncological and Immunological Effects of

321

to 29 days post-treatment (Fig.5B) and general survival by 8 days from 43 days to 51

days post-treatment (Fig.5C).

8.5.3 Histotripsy Ablation Results in Increased Acute Cellular Immune Response

Figure 8-6 Analysis of mRNA expression in treated tumors showed regulation of immune pathways (A). Regulated pathways interact with each other and are up regulated in the acute group and downregulated in the chronic group (B). Serum HMBG-1 levels (C) correlate with the mRNA upregulation of HMGB-1 associated pathways.

Page 359: Determining the Oncological and Immunological Effects of

322

These tumors even without treatment tend to develop a central area of necrosis as

they progress. These necrotic cores are often characterized by accumulations of cell

debris with variable numbers of infiltrating degenerate and nondegenerate neutrophils.

Otherwise, appreciable numbers of immune cells are relatively absent microscopically

throughout the tumor tissue (Fig.6A). Treated tumors also exhibited a core of cell death

following ablation with variable infiltration by predominantly degenerate neutrophils.

However, in addition to cellular debris, these cores also often contain ghost cells and

acute hemorrhage (Fig.6B). In both treated and untreated tumors, small to moderate

numbers of neutrophils, macrophages, lymphocytes, and/or plasma ells are present at

the tumor periphery. There is no appreciable difference between the two (Fig.6C-D).

Despite the lack of appreciable differences in immune cell populations microscopically,

we investigated immune cell signaling through gene expression. Super array rtPCR

showed many genes associated with cancer inflammation and immunity crosstalk were

significantly regulated after histotripsy treatment (Sup Table 1). IPA analysis comparing

treated animals to untreated in acute and survival groups found multiple canonical

immune pathways regulated by histotripsy treatment (Fig.6A). As a trend, the pro-

inflammatory pathways are upregulated 24 hours treatment, but the majority of these

pathways become downregulated and were replaced with more anti-inflammatory

pathways at survival points. Many of the upregulated pathways (HMGB1, NF-κB, IL-6,

and TLR signaling pathways) are interconnected and are self-regulated to decrease in

function over time (Fig.6B). The schematic in Figure 5 illustrates the interactions and

mechanisms identified by IPA that are upregulated in the acute treatment group and

downregulated in the survival group. Some of these proteins and pathways were not

Page 360: Determining the Oncological and Immunological Effects of

323

directly analyzed, but are predicted to be modulated based upon molecules that are

upstream and downstream by IPA. Together, these data indicate a significant

upregulation of pathways associated with the activation of the innate immune system

associated with DAMP signaling.

HMGB1 signaling was identified by IPA and is a potent DAMP. To verify this aspect

of our pathway analysis, we evaluated the protein levels of HMGB1 in the serum with

and without histotripsy treatment. Consistent with the IPA results, HMGB1 levels were

increased in the sera of treated mice at the acute time point compared to untreated and

control animals, while both treated and untreated mice saw a significant (p<0.05)

decrease in serum HMGB1 levels at survival endpoints (Fig.6C). Although there was a

notable increase, due to the significantly higher variance (p=0.0032), there was no

significance in the average serum HMGB1 level in the treated acute group compared to

the untreated and control groups.

8.5.4 Histotripsy Alters Immune Cell Composition in the Tumor Microenvironment

To quantify changes in immune cell populations within tumors over time after

histotripsy treatment, tumors were collected 24 hours, 7 days, and 14 days after

treatment for flow cytometry. Although there were no significant changes at the 24-hour

time point, there was an appreciable decrease in inflammatory Dendritic cells (iDCs,

p=0.19), classical Dendritic cells (cDCs, p=0.18), granulocytes, Th cells, CD4+ T cells,

and CD8+ T cell populations after treatment (Fig.6-7). The relative reduction of iDCs

(p=0.13) and cDCs (p=0.07) continued to day 7 along with a decrease in M-MDSCs, at

Page 361: Determining the Oncological and Immunological Effects of

324

this point the remaining cells that appeared reduced at 24 hours were more comparable

to the untreated (Fig.7). At 14 days after treatment, macrophages and Treg cells were

found to be significantly reduced while cDCs were found to be significantly increased

and neutrophils (p=0.19) notably increased in treated tumors compared to the untreated

(Fig.7-8). While there we no changes in the ratios of CD4+/CD8+ T cells in the treated

tumors compared to the untreated, there were significant increases in CD4+ and a

notable increase in CD8+ T cells within the treated tumors at 7 days after treatment

(Fig.8).

Figure 8-7 Single cell suspension from treated and untreated tumors were collected at 24 hours, 7 days, and 14 days after treatment and were stained as described for flow cytometry to identify innate immune cells. Percentage of each innate immune cell population analyzed was calculated as part of the total CD45+ cells stained. Example flow cytometry plots from treated and untreated tumors at 14 days after treatment.

Page 362: Determining the Oncological and Immunological Effects of

325

Figure 8-8 Single cell suspension from treated and untreated tumors were collected at 24 hours, 7 days, and 14 days after treatment and were stained as described for flow cytometry to identify adaptive immune cells. Percentage of each adaptive immune cell population analyzed was calculated as part of the total viable cells stained (gated as singlets). The ratio of CD4+/CD8+ T cells was calculated as a simple ratio. Example flow cytometry plots from treated and untreated tumors at 14 days after treatment.

8.6 Discussion

This study investigated the treatment of pancreatic tumors with histotripsy and

the resultant immunological responses, with a focus on the innate immune system. To

optimize our immune system assessments, we established conservative margins with

the expectation that some tumor would not be treated. This allowed cells within the

untreated margins to respond to the effects of our treatment and provided specimens for

Page 363: Determining the Oncological and Immunological Effects of

326

subsequent analysis. For in vivo ablations, histotripsy treatment was confirmed via

bubble-cloud formation, while H&E showed complete ablation within targeted regions.

Overall, we achieved a partial ablation of all treated tumors. There was an average

reduction of 43% in the tumor diameters of the remaining tumor tissue after treatment,

comparable to tumor retardation compared to previous histotripsy subcutaneous tumor

treatments [20, 23, 24]. Based upon the fully ablated tissue found within the targeted

regions on H&E, the remaining tissue can be assumed to have been untreated based

upon the margins set. This led to tumors that were fully ablated in certain regions and

completely untreated in others. More complete ablation of tumors with histotripsy has

been achieved in de novo and in situ studies where margins include surrounding

healthy tissues, but these tumors still recurred [40-43]. In this study, the progression of

tumor growth was improved with treatment (Fig.5B). However, in line with previous

studies, tumor recurrence occurred and minimized the improvement of the general

survival for animals that received treatment (Fig.5C).

The release of DAMPs is a consistent feature of focal ablation modalities, and

the established modalities have been compared to each other in previous work [16].

Histotripsy has not been previously, directly compared to non-ultrasonic ablation

modalities. The effects of histotripsy compared to other ablation modalities for producing

immune-stimulating molecules is of interest given the response to the histotripsy

treatment of pancreatic tumors in vivo yielded an inflammatory response that is turned

off over time while showing relatively large variations of immune cell proportions within

treated tumors. Looking at the release of DNA and peptides as potential DAMPs,

Page 364: Determining the Oncological and Immunological Effects of

327

showed that histotripsy is comparable to the non-thermal ablations cryoablation and IRE

(Fig.3).

Extracellular DNA is recognized by innate immune system receptors as a DAMP

and is not typically found in the absence of damage [44]. Additionally, the presence of

released proteins increases the probability of the immune system being recruited by

damage associated cytokines and antigens. Our hypothesized expectations for the in

vitro studies were that cryoablation and histotripsy should have similar, more intact

DNA, in larger fragments, and more protein released from cells after ablation given that

they both ablate cells through immediate lysis [17, 45]. On the other hand, no cell death

was observed in untreated cells, also as expected, and therefore had low levels of

protein and DNA. However, IRE induces a delayed cell death, defined as either

apoptosis or pyroptosis [15]. DNA fragmentation and protein release or damage are

hallmark features of programed cell death. Similarly, thermal ablation is known to

directly denature DNA and protein [16, 46]. Thus, in both of these cases, we predicted

that the DNA released following ablation would be significantly more fragmented,

including significantly smaller and undetectable fragments. Even though IRE does use a

delayed cell death mechanism, we did expect to find the higher level of detectable

proteins given that programmed cell death does not denature all of the released

proteins and previous studies that also showed that IRE can release a significant

magnitude of intact proteins [15, 16].

In addition, results from the in vitro experiments provide important comparisons

between different ablation methods and demonstrate that non-thermal ablation

approaches, including histotripsy, lead to significantly increased release of DAMPs and

Page 365: Determining the Oncological and Immunological Effects of

328

potential antigens in comparison to thermal ablation methods. In comparing the clinical

relevance of the differential release of DAMPs and potential antigens that was observed

between the ablation modalities in vitro, it is important to note that additional in vivo

studies will be needed to further study the differences between each ablation modality.

Similarly, additional in vivo studies will be needed to further investigate the role of

treatment dose in immune system activation for each of the ablation modalities. For

instance, it is not clear whether the partial ablations generated in our in vitro studies will

be representative of a partial or incomplete ablation in a clinical setting. For the thermal

ablation, cryoablation, and IRE samples treated at partial-ablation doses in this study in

vitro, all of the cells in suspension were treated at a reduced magnitude. In contrast,

since histotripsy is a binary treatment that requires a bubble cloud to be formed once a

distinct pressure threshold has been exceeded [47-50], the histotripsy partial ablation

resulted in only a portion of the cells being exposed to a full amplitude histotripsy bubble

cloud, whereas the remaining cells received no treatment. When comparing these in

vitro results from cell suspensions to in vivo ablations, it is important to remember that

these differences in how a partial ablation versus a full ablation is acquired may result in

different responses. As a result, controlled in vivo studies comparing the effects of

different ablation modalities as well as different treatment doses on the potential

immunological benefits should be conducted. In addition to comparing the differences

between partial and complete ablation, these future studies should investigate the

potential risks of overtreatment that could potentially reduce immunological benefits. For

instance, in prior work designing IRE protocols, users have outlined specific treatment

guidelines to avoid excessive Joule heating to capitalize on the therapeutic benefits of

Page 366: Determining the Oncological and Immunological Effects of

329

the non-thermal IRE ablation [34]. Similarly, samples over treated with thermal ablation

modalities and cryoablation face an increased rate of macromolecule breakdown and

protein denaturation compared to non-thermal [16]. Future studies are needed to

determine any effects of overtreatment with histotripsy and to determine the optimal

histotripsy treatment strategies for maximizing immunological benefits.

Given that histotripsy’s mechanism of ablation is the mechanical lysis of cells and

that in vitro we found increased release of DNA and proteins (Fig.3), the determination

that DAMP signaling pathways (Fig.5F) were activated in vivo was not surprising.

Although we did not appreciate histological changes in immune cell infiltration on a

microscopic level, gene expression analysis suggested changes in immune cell

signaling pathways which led us to assess immune cell populations via flow cytometry,

a more sensitive indicator. Here we were able to identify specific pathways modulated

by histotripsy, including HMGB1, IL-6, Interferon, TEC Kinase, JAK/STAT, and PPAR

pathway signaling. These pathways are consistent with responses to trauma or physical

damage [51-53]. As time passes after a trauma, these signaling pathways become

repressed to control the immune response. One way this is done is through the PPAR

pathway, which can inhibit the downstream production of cytokines from the HMGB1

and NF-κB pathways [54]. The activation of the PPAR pathway can also increase

healing effects by stimulating the production of VEGF [55], found in our study to be

significantly increased in treated survival group tumors (Fig.5F, Sup Table 1). For

healing, the production of VEGF is needed to reestablish healthy vasculature; however,

high levels of VEGF within a tumor have been established to be tumorigenic and

correlates with poor patient outcomes [56]. Histotripsy increasing the activation of the

Page 367: Determining the Oncological and Immunological Effects of

330

PPAR pathway and increasing the levels of VEGF expression did not promote negative

outcomes for the animals in our study but could be targets of future adjuvants to extend

the early inflammatory tumor microenvironment.

At two weeks post-histotripsy ablation, there was a significant decrease in

macrophages (Fig.6) and Treg cells (Fig.7). Significant decreases in the expression of

cytokines associated with tumor-associated macrophages (TAMs) including IL-4, IL-10

CCL-2, CCL-22, and CXCl-12 (Sup. Table 1) and suggest that the reduction in

macrophages within the treated tumors could be indicative of a decrease in TAMs [57,

58]. A decrease in Treg cells with a potential decrease in TAMs could indicate additional

access of histotripsy immunomodulation of the tumor microenvironment to being less

anti-inflammatory. This could be a potential target for enhancement with adjuvant

therapies [57, 59]. Overall, these changes to immune cell populations should be further

analyzed in future studies to determine the full extent of changes to sub-populations,

such as changes to M1/M2/TAM macrophages instead of basic populations shown

here.

Early studies investigating the immunological effects of histotripsy compared the

effects of acoustic cavitation to acoustic heating with thermal HIFU. One study using a

subcutaneous colon adenocarcinoma mouse model showed that histotripsy is capable

of stimulating CD11c+ cells within the tumors more than thermal HIFU [26]. It has also

been shown that histotripsy of murine melanoma can better stimulate an immune

response with a decrease in metastasis in the weeks following treatment compared to

HIFU, suggesting involvement of anti-tumor lymphocytes [27]. More recent studies have

further established the pro-inflammatory local and systemic effects of histotripsy on the

Page 368: Determining the Oncological and Immunological Effects of

331

immune response in melanoma, neuroblastoma, hepatocellular carcinoma, and renal

cell carcinoma [22-24]. This study adds to this knowledge by establishing a framework

for the immune response to histotripsy ablation of pancreatic cancer. The response

reestablished here is similar to other studies. However, given that the Pan02 tumors are

known to be poorly immunogenic [60], it was not surprising that the local changes in

immune cell populations, while significant at points (Figs.7-8), were not as prominent of

a profile shift as what has been reported in other tumors types. Using the Pan02 model,

a prior study with IRE showed that subcutaneous tumors did not have a strong of a

change in immune cell populations within the tumor as orthotopic tumors [61]. Similar to

the current work showed data that non-thermal ablation of the subcutaneous pancreatic

tumors can shift the tumor microenvironment to being more pro-inflammatory [61].

The results of this study provide evidence that histotripsy can ablate subcutaneous

pancreatic tumors and stimulate a local immune response. This study builds upon

previous studies utilizing histotripsy for other tumor types [20, 22-24], and shows a

potential immune profile for pancreatic tumors after histotripsy ablation. Overall, the

results of this work provide a baseline expectation of the response of pancreatic tumors

to histotripsy, which will help for planning future orthotopic studies.

8.7 Conclusion

This study demonstrates the feasibility of using histotripsy for pancreatic cancer

ablation and defines mechanisms associated with innate immune system activation

Page 369: Determining the Oncological and Immunological Effects of

332

following treatment. Further work is needed to establish the mechanisms behind the

immunomodulation of the tumor microenvironment and systemic effects.

8.8 Acknowledgment

This work was supported by the Virginia Maryland College of Veterinary Medicine; The

Virginia Tech Institute for Critical Technology and Applied Sciences Center for

Engineered Health; and The National Institutes of Health. The content is solely the

responsibility of the authors and does not necessarily represent the official views of the

NIH or any other funding agency. Figures 1B and 6F were created using biorender.com.

Page 370: Determining the Oncological and Immunological Effects of

333

8.9 References

[1] R. L. Siegel, K. D. Miller, and A. Jemal, "Cancer statistics, 2017," CA: a cancer

journal for clinicians, vol. 67, no. 1, pp. 7-30, 2017.

[2] R. L. Siegel, K. D. Miller, and A. Jemal, "Cancer statistics, 2020," CA: a cancer

journal for clinicians, vol. 70, no. 1, pp. 7-30, 2020.

[3] H. Manuel, "Pancreatic cancer," New England Journal of Medicine, vol. 362, no.

17, 2010.

[4] S. Gillen, T. Schuster, C. M. Zum Büschenfelde, H. Friess, and J. Kleeff,

"Preoperative/neoadjuvant therapy in pancreatic cancer: a systematic review and

meta-analysis of response and resection percentages," PLoS medicine, vol. 7,

no. 4, 2010.

[5] O. Kepp, A. Marabelle, L. Zitvogel, and G. Kroemer, "Oncolysis without viruses—

inducing systemic anticancer immune responses with local therapies," Nature

Reviews Clinical Oncology, pp. 1-16, 2019.

[6] C. J. Simon, D. E. Dupuy, and W. W. Mayo-Smith, "Microwave ablation:

principles and applications," Radiographics, vol. 25, no. suppl_1, pp. S69-S83,

2005.

[7] Y. Keisari, "Tumor abolition and antitumor immunostimulation by physico-

chemical tumor ablation," Front Biosci (Landmark Ed), vol. 22, pp. 310-47, 2017.

[8] R. V. Davalos, L. Mir, and B. Rubinsky, "Tissue ablation with irreversible

electroporation," Annals of biomedical engineering, vol. 33, no. 2, p. 223, 2005.

Page 371: Determining the Oncological and Immunological Effects of

334

[9] R. C. Martin et al., "Treatment of 200 locally advanced (stage III) pancreatic

adenocarcinoma patients with irreversible electroporation: safety and efficacy,"

Annals of surgery, vol. 262, no. 3, pp. 486-494, 2015.

[10] H. J. Scheffer et al., "Ablation of locally advanced pancreatic cancer with

percutaneous irreversible electroporation: results of the phase I/II PANFIRE

study," Radiology, vol. 282, no. 2, pp. 585-597, 2017.

[11] M. P. Belfiore et al., "Percutaneous CT-guided irreversible electroporation

followed by chemotherapy as a novel neoadjuvant protocol in locally advanced

pancreatic cancer: our preliminary experience," International Journal of Surgery,

vol. 21, pp. S34-S39, 2015.

[12] H. J. Scheffer et al., "Irreversible electroporation of locally advanced pancreatic

cancer transiently alleviates immune suppression and creates a window for

antitumor T cell activation," Oncoimmunology, vol. 8, no. 11, p. 1652532, 2019.

[13] R. C. Martin et al., "Irreversible electroporation in locally advanced pancreatic

cancer: a call for standardization of energy delivery," Journal of Surgical

Oncology, vol. 114, no. 7, pp. 865-871, 2016.

[14] N. Beitel-White, R. C. G. Martin, Y. Li, R. M. Brock, I. C. Allen, and R. V.

Davalos, "Real-time prediction of patient immune cell modulation during

irreversible electroporation therapy," (in eng), Sci Rep, vol. 9, no. 1, p. 17739,

Nov 28 2019, doi: 10.1038/s41598-019-53974-w.

[15] R. M. Brock, N. Beitel-White, R. V. Davalos, and I. C. Allen, "Starting a Fire

Without Flame: The Induction of Cell Death and Inflammation in Electroporation-

Based Tumor Ablation Strategies," Frontiers in Oncology, vol. 10, 2020.

Page 372: Determining the Oncological and Immunological Effects of

335

[16] Q. Shao et al., "Engineering T cell response to cancer antigens by choice of focal

therapeutic conditions," International Journal of Hyperthermia, vol. 36, no. 1, pp.

130-138, 2019.

[17] K. B. Bader, E. Vlaisavljevich, and A. D. Maxwell, "For whom the bubble grows:

physical principles of bubble nucleation and dynamics in histotripsy ultrasound

therapy," Ultrasound in medicine & biology, 2019.

[18] E. Vlaisavljevich, A. Maxwell, L. Mancia, E. Johnsen, C. Cain, and Z. Xu,

"Visualizing the Histotripsy Process: Bubble Cloud-Cancer Cell Interactions in a

Tissue-Mimicking Environment," (in Eng), Ultrasound Med Biol, Jul 8 2016, doi:

S0301-5629(16)30099-0 [pii] 10.1016/j.ultrasmedbio.2016.05.018.

[19] E. Vlaisavljevich et al., "Non-invasive ultrasound liver ablation using histotripsy:

Chronic study in an in vivo rodent model," Ultrasound in medicine & biology, vol.

42, no. 8, pp. 1890-1902, 2016.

[20] T. Worlikar et al., "Histotripsy for Non-Invasive Ablation of Hepatocellular

Carcinoma (HCC) Tumor in a Subcutaneous Xenograft Murine Model," in 2018

40th Annual International Conference of the IEEE Engineering in Medicine and

Biology Society (EMBC), 2018: IEEE, pp. 6064-6067.

[21] K. C. Longo et al., "Robotically Assisted Sonic Therapy (RAST) for Noninvasive

Hepatic Ablation in a Porcine Model: Mitigation of Body Wall Damage with a

Modified Pulse Sequence," Cardiovasc Intervent Radiol, vol. 42, no. 7, pp. 1016-

1023, Jul 2019, doi: 10.1007/s00270-019-02215-8.

[22] G. R. Schade, Y.-N. Wang, S. D'Andrea, J. H. Hwang, W. C. Liles, and T. D.

Khokhlova, "Boiling Histotripsy Ablation of Renal Cell Carcinoma in the Eker Rat

Page 373: Determining the Oncological and Immunological Effects of

336

Promotes a Systemic Inflammatory Response," Ultrasound in medicine &

biology, vol. 45, no. 1, pp. 137-147, 2019.

[23] A. Eranki et al., "High-Intensity Focused Ultrasound (HIFU) Triggers Immune

Sensitization of Refractory Murine Neuroblastoma to Checkpoint Inhibitor

Therapy," Clinical Cancer Research, vol. 26, no. 5, pp. 1152-1161, 2020.

[24] S. Qu et al., "Non-thermal histotripsy tumor ablation promotes abscopal immune

responses that enhance cancer immunotherapy," Journal for ImmunoTherapy of

Cancer, vol. 8, no. 1, 2020.

[25] K. J. Pahk et al., "Boiling Histotripsy-induced Partial Mechanical Ablation

Modulates Tumour Microenvironment by Promoting Immunogenic Cell Death of

Cancers," Scientific reports, vol. 9, no. 1, pp. 1-12, 2019.

[26] Z. Hu et al., "Investigation of HIFU-induced anti-tumor immunity in a murine

tumor model," Journal of translational medicine, vol. 5, no. 1, p. 34, 2007.

[27] Y. Xing, X. Lu, E. C. Pua, and P. Zhong, "The effect of high intensity focused

ultrasound treatment on metastases in a murine melanoma model," Biochemical

and biophysical research communications, vol. 375, no. 4, pp. 645-650, 2008.

[28] M. E. Lorkowski et al., "Immunostimulatory nanoparticle incorporating two

immune agonists for the treatment of pancreatic tumors," (in eng), J Control

Release, Nov 11 2020, doi: 10.1016/j.jconrel.2020.11.014.

[29] C. C. Baniel et al., "In situ Vaccine Plus Checkpoint Blockade Induces Memory

Humoral Response," (in eng), Front Immunol, vol. 11, p. 1610, 2020, doi:

10.3389/fimmu.2020.01610.

Page 374: Determining the Oncological and Immunological Effects of

337

[30] A. D. Maxwell, C. A. Cain, A. P. Duryea, L. Yuan, H. S. Gurm, and Z. Xu,

"Noninvasive thrombolysis using pulsed ultrasound cavitation therapy–

histotripsy," Ultrasound in medicine & biology, vol. 35, no. 12, pp. 1982-1994,

2009.

[31] B. Aarts et al., "Cryoablation and immunotherapy: an overview of evidence on its

synergy," Insights into imaging, vol. 10, no. 1, pp. 1-12, 2019.

[32] C. Brace, "Thermal tumor ablation in clinical use," (in eng), IEEE Pulse, vol. 2,

no. 5, pp. 28-38, Sep-Oct 2011, doi: 10.1109/mpul.2011.942603.

[33] M. Faroja et al., "Irreversible electroporation ablation: is all the damage

nonthermal?," Radiology, vol. 266, no. 2, pp. 462-470, 2013.

[34] R. V. Davalos, S. Bhonsle, and R. E. Neal, "Implications and considerations of

thermal effects when applying irreversible electroporation tissue ablation

therapy," The Prostate, vol. 75, no. 10, pp. 1114-1118, 2015.

[35] R. M. Brock et al., "Patient derived xenografts expand human primary pancreatic

tumor tissue availability for ex vivo irreversible electroporation testing," Frontiers

in Oncology, vol. 10, 2020.

[36] V. M. Ringel-Scaia et al., "High-frequency irreversible electroporation is an

effective tumor ablation strategy that induces immunologic cell death and

promotes systemic anti-tumor immunity," EBioMedicine, vol. 44, pp. 112-125, Jun

2019, doi: 10.1016/j.ebiom.2019.05.036.

[37] M. Kato, K. Watabe, M. Tsujii, T. Funahashi, I. Shimomura, and T. Takehara,

"Adiponectin inhibits murine pancreatic cancer growth," Digestive diseases and

sciences, vol. 59, no. 6, pp. 1192-1196, 2014.

Page 375: Determining the Oncological and Immunological Effects of

338

[38] J. I. Youn and D. I. Gabrilovich, "The biology of myeloid‐derived suppressor cells:

the blessing and the curse of morphological and functional heterogeneity,"

European journal of immunology, vol. 40, no. 11, pp. 2969-2975, 2010.

[39] S. Rose, A. Misharin, and H. Perlman, "A novel Ly6C/Ly6G‐based strategy to

analyze the mouse splenic myeloid compartment," Cytometry Part A, vol. 81, no.

4, pp. 343-350, 2012.

[40] N. R. Styn, T. L. Hall, J. B. Fowlkes, C. A. Cain, and W. W. Roberts, "Histotripsy

of renal implanted VX-2 tumor in a rabbit model: investigation of metastases,"

Urology, vol. 80, no. 3, pp. 724-729, 2012.

[41] G. R. Schade et al., "Histotripsy focal ablation of implanted prostate tumor in an

ACE-1 canine cancer model," The Journal of urology, vol. 188, no. 5, pp. 1957-

1964, 2012.

[42] T. Worlikar et al., "Non-Invasive Orthotopic Liver Tumor Ablation Using

Histotripsy in an in Vivo Rodent Hepatocellular Carcinoma (HCC) Model,"

presented at the Society of Interventional Oncology, Boston, MA, USA, 2019.

[43] J. Vidal Jove et al., "Phase I Study of Safety and Efficacy of Hepatic Histotripsy:

Preliminary Results of First in Man Experience with Robotically-Assisted Sonic

Therapy," presented at the International Society of Therapeutic Ultrasound,

Barcelona, Spain, 2019.

[44] D. S. Pisetsky, "The origin and properties of extracellular DNA: from PAMP to

DAMP," Clinical Immunology, vol. 144, no. 1, pp. 32-40, 2012.

Page 376: Determining the Oncological and Immunological Effects of

339

[45] J. P. Erinjeri and T. W. Clark, "Cryoablation: mechanism of action and devices,"

Journal of Vascular and Interventional Radiology, vol. 21, no. 8, pp. S187-S191,

2010.

[46] K. F. Chu and D. E. Dupuy, "Thermal ablation of tumours: biological mechanisms

and advances in therapy," Nature Reviews Cancer, vol. 14, no. 3, pp. 199-208,

2014.

[47] E. Vlaisavljevich, T. Gerhardson, T. Hall, and Z. Xu, "Effects of f-number on the

histotripsy intrinsic threshold and cavitation bubble cloud behavior," Phys Med

Biol, vol. 62, no. 4, pp. 1269-1290, Feb 21 2017, doi: 10.1088/1361-

6560/aa54c7.

[48] E. Vlaisavljevich et al., "Effects of Ultrasound Frequency and Tissue Stiffness on

the Histotripsy Intrinsic Threshold for Cavitation," Ultrasound Med Biol, 2015.

[49] E. Vlaisavljevich, A. Maxwell, M. Warnez, E. Johnsen, C. A. Cain, and Z. Xu,

"Histotripsy-induced cavitation cloud initiation thresholds in tissues of different

mechanical properties," IEEE Trans Ultrason Ferroelectr Freq Control, vol. 61,

no. 2, pp. 341-52, Feb 2014, doi: 10.1109/TUFFC.2014.6722618.

[50] A. D. Maxwell, C. A. Cain, T. L. Hall, J. B. Fowlkes, and Z. Xu, "Probability of

cavitation for single ultrasound pulses applied to tissues and tissue-mimicking

materials," Ultrasound in medicine & biology, vol. 39, no. 3, pp. 449-465, 2013.

[51] D. S. Pisetsky, "Cell death in the pathogenesis of immune-mediated diseases:

the role of HMGB1 and DAMP-PAMP complexes," Swiss medical weekly, vol.

141, p. w13256, 2011.

Page 377: Determining the Oncological and Immunological Effects of

340

[52] E. Venereau, M. Schiraldi, M. Uguccioni, and M. E. Bianchi, "HMGB1 and

leukocyte migration during trauma and sterile inflammation," Molecular

immunology, vol. 55, no. 1, pp. 76-82, 2013.

[53] N. Haque et al., "Role of the CXCR4-SDF1-HMGB1 pathway in the directional

migration of cells and regeneration of affected organs," World Journal of Stem

Cells, vol. 12, no. 9, p. 938, 2020.

[54] S. Ying, X. Xiao, T. Chen, and J. Lou, "PPAR ligands function as suppressors

that target biological actions of HMGB1," PPAR research, vol. 2016, 2016.

[55] L. Piqueras et al., "Activation of PPARβ/δ induces endothelial cell proliferation

and angiogenesis," Arteriosclerosis, thrombosis, and vascular biology, vol. 27,

no. 1, pp. 63-69, 2007.

[56] A. Lapeyre-Prost et al., "Immunomodulatory activity of VEGF in cancer," in

International review of cell and molecular biology, vol. 330: Elsevier, 2017, pp.

295-342.

[57] J. Zhou, Z. Tang, S. Gao, C. Li, Y. Feng, and X. Zhou, "Tumor-Associated

Macrophages: Recent Insights and Therapies," Frontiers in Oncology, vol. 10, p.

188, 2020.

[58] G. J. Szebeni, C. Vizler, K. Kitajka, and L. G. Puskas, "Inflammation and cancer:

extra-and intracellular determinants of tumor-associated macrophages as tumor

promoters," Mediators of inflammation, vol. 2017, 2017.

[59] B. Chaudhary and E. Elkord, "Regulatory T cells in the tumor microenvironment

and cancer progression: role and therapeutic targeting," Vaccines, vol. 4, no. 3,

p. 28, 2016.

Page 378: Determining the Oncological and Immunological Effects of

341

[60] A. Kumar, K. Chamoto, P. S. Chowdhury, and T. Honjo, "Tumors attenuating the

mitochondrial activity in T cells escape from PD-1 blockade therapy," ELife, vol.

9, p. e52330, 2020.

[61] C. He, X. Huang, Y. Zhang, X. Lin, and S. Li, "T‐cell activation and immune

memory enhancement induced by irreversible electroporation in pancreatic

cancer," Clinical and Translational Medicine, vol. 10, no. 2, p. e39, 2020.

Page 379: Determining the Oncological and Immunological Effects of

342

8.10 Supplemental Table and Figure Legends

Figure 8-9 Supplemental Figure 1: Whole western blot images analyzing HA antigen released in ablated cell supernatants.

Page 380: Determining the Oncological and Immunological Effects of

343

Table 8-3 Supplemental Table 1: Genes analyzed from SuperArry rtPCR and calculated fold regulation.

Page 381: Determining the Oncological and Immunological Effects of

344

Chapter 9. Conclusion and Future Directions

Page 382: Determining the Oncological and Immunological Effects of

345

9.1 Introduction

This dissertation consists of three parts. Part One of this dissertation (Chapters 2- 3)

focuses on determining the safety and dosages for using histotripsy to treat stronger

hepatopancreatobiliary tumors using excised tissues and patient derived xenograft

(PDX) mice. Part Two (Chapters 4-6) moves onto more complex models, using large

animals and veterinary patients, to establish application protocols for additional tumor

types. Part Three (Chapters 7-8) shifts to utilizing immunocompetent mouse models to

investigate the immunological effects of histotripsy in pancreatic adenocarcinoma.

9.2 How can we safely and efficacious ablate more difficult tumors?

In Chapter 2, the role of histotripsy dose in the susceptibility of various liver

tumors is investigated. Specifically, it was determined that while using a 700KHz

histotripsy transducer with a PRF of 500Hz, hepatocellular carcinoma tumors can be

fully ablated with an applied dose 500 pulses/point (Fig.2-5). However, when looking at

cholangiocarcinoma (CC) tumors, known to be highly fibrotic and have a higher Young’s

modulus than most other liver malignancies [REF], even at a dosage of 4000

pulses/point there was about 25% cell viability within the targeted area (Fig.2-7E).

Even though this ex vivo study was limited by sample numbers, due to the rarity of

CC, a patient-derived xenograft (PDX) tumor model of CC was utilized to determine if it

could be safe and practical to achieve a significant ablation with histotripsy. In two

Page 383: Determining the Oncological and Immunological Effects of

346

different PDX cohorts, with tumors derived from different patients, it was found that

using a small, 8-element 1MHz transducer at a PRF of 500Hz, that a complete ablation

of targeted tumor was achieved at 500 pulses/point (Fig.2-3K and Fig.3-2B). Knowing

that it was feasible to ablate the PDX CC tumors with a reasonable dose, Chapter 3

focused on determining whether ablation of the full tumor volume is safe. When

applying a 1-2mm positive margin to ensure the application of histotripsy to the entire

CC tumor, the off-target damage was deemed too great to justify (Fig.3-3E-F).

However, when applying a 0mm margin, therefore attempting to target 100% of the

tumor and no margin, the ablation safety was significantly higher, leading to the only

adverse events seen being mild bruising immediately after treatment and a small scab

within the first week (Fig.3-3D). Within 2.5 weeks of treatment, as the ablated

homogenate was resorbed, the decrease in tumor diameter was found to reach a

maximum of 73%, when compared to the untreated tumors, including the temporary

disappearance of 3 of the 6 treated tumors (Fig.3-4). Future work for CC tumor ablation

needs to determine how to utilize an effective ablation dose on the tumor to achieve a

full ablation without causing significant damage to off target tissues.

9.3 Can we use a large animal model to develop histotripsy for acoustically

difficult to target tumor locations?

As discussed in Chapter 4, testing histotripsy devices in rodents requires smaller,

custom made therapeutic systems (Fig.4-1), while utilizing large animal models and

veterinary patients allows for the testing of human scale systems (Fig.4-2). Pilot studies

Page 384: Determining the Oncological and Immunological Effects of

347

of canine soft tissue sarcoma and osteosarcoma demonstrate the feasibility of utilizing

these de novo tumor carrying animals to determine therapeutic protocols for histotripsy

(Fig.4-7,8).

While there are many benefits to using veterinary patients, one drawback is the

lack of easy replication of exact tumor size, location, and severity that is offered by

tumor models. Chapters 5 and Appendix A lay the foundation for our newly

established porcine tumor model. By using CRISPR/Cas9 genetically modified pigs that

lack function RAG2 and IL-2RG proteins (Fig.6-1), which led the pigs to be severely

immunocompromised (Fig.6-2), a range of established human and murine cell lines can

be engrafted for tumor generation (Fig.4-4, 5-3, A-2). Focusing investigations on

pancreatic cancer, the engraftment of the human pancreatic adenocarcinoma cell line,

Panc01, showed luminal structures (Fig.5-5) and expression of the cytokeratin protein

CK-19 (Fig.5-6) that is consistent with human pancreatic tumors. While this study

established a porcine model for pancreatic cancer, before using these animals it is

necessary to establish a safe and effective protocol for targeting the pancreas with

histotripsy.

Chapter 6 investigates the potential of using histotripsy to target the porcine

pancreas. The goal of this project was to determine the safety of the treatment as well

as establishing the least invasive protocol for using histotripsy. The first pilot study

demonstrated that with minimal intestinal gas reduction (i.e. fasting for 12 hours)

resulted in poor acoustic visualization of the pancreas (Fig.6-3B,4) and mild damage to

the intestines (Fig.6-4). For subsequent studies, the animals were fed a custard that

contained laxative and simethicone, the combination of which allowed for the clear

Page 385: Determining the Oncological and Immunological Effects of

348

visualization of the pancreas (Fig.6-3C) and histotripsy ablation (Fig.6-5). Using this

protocol, 4 animals were survived one week with no negative side effects noted in

behavior, diet, blood work (Fig.6-7), CT imaging (Fig.6-8), or necropsy (Fig.6-6,9). This

study established a protocol for safely targeting the pancreas for ablation with

histotripsy. Future work needs to determine the feasibility of this protocol for targeting

tumors within the pancreas.

9.4 How do different tumor types immunologically respond to histotripsy?

Chapter 7 establishes the foundation of histotripsy’s role in immunomodulation.

This literature review establishes the pre-clinical immune effects caused by the

mechanical ablation of tumors with histotripsy. This includes production of damage

associated molecular patterns, release of immune stimulating cytokines, changes in

local immune cell populations to those that are more pro-inflammatory and anti-tumor,

and increases in systemic immunity (Fig.7-5). However, this review also serves to

highlight future work that is still needed, including investigations into repolarization of

pro-tumor immune cells, synergistic effects of check-point inhibitors and other

chemotherapeutics, and the full extent of systemic, anti-body mediated immunity

stimulated against the tumor. In sum, these studies suggest that there is a reproducible,

consistent, and perhaps even tunable immune effect generated by histotripsy modalities

that is consistent across multiple tumor types. This review also highlights that all of this

work has been done utilizing small animal models and only across a few tumor types. In

order to fully understand the scope of the immunological effects of histotripsy, more

Page 386: Determining the Oncological and Immunological Effects of

349

work needs to be done in additional tumor types, large animals, and human clinical

trials. Additionally, future work should investigate how histotripsy, a non-thermal and

non-invasive, ablation’s effects compare to other, established ablation modalities.

Following this, Chapter 8 focused on determining the potential immune effects of

histotripsy on pancreatic cancer using the murine Pan02 model. To study the effects of

histotripsy on pancreatic cancer, we utilized an in vitro model of pancreatic

adenocarcinoma and compared the release of neoantigens following histotripsy

treatment to other ablation modalities (Fig.8-2). Histotripsy was found to release

immune-stimulating molecules at magnitudes similar to other non-thermal ablation

modalities and superior to thermal ablation modalities, which corresponded to increased

innate immune system activation in vivo (Fig.8-3). In subsequent in vivo studies, murine

Pan02 tumors were treated with histotripsy, and observed significantly increased

progression-free and overall survival (Fig.8-5), with increased activation of the innate

immune system 24 hours post-treatment (Fig.8-6) and decreased tumor-associated

immune cell populations within 14 days of treatment (Fig.8-7,8). This study

demonstrates the feasibility of using histotripsy for pancreatic cancer ablation and

provides mechanistic insight into the initial innate immune system activation following

treatment. Further work is needed to establish the mechanisms behind the

immunomodulation of the tumor microenvironment and systemic effects.

Page 387: Determining the Oncological and Immunological Effects of

350

9.5 Overall Conclusion

The results of this dissertation provide insight into establishing protocols for treating

new types of tumors with histotripsy and immunological effects that lay groundwork for

improving future co-therapeutic planning. Future work will aim to translate histotripsy

into clinical applications and determining co-therapies that can improve local and

systemic tumor burden.

Page 388: Determining the Oncological and Immunological Effects of

351

Appendix A. Employing Novel Porcine Models of Subcutaneous Pancreatic Cancer to Evaluate Oncological Therapies

Alissa Hendricks-Wenger, Margaret A Nagai-Singer, Kyungjun Uh, Eli Vlaisavljevich, Kiho Lee, Irving C Allen

This chapter is excerpted from a textbook chapter accepted to “Bioengineering

Technology,” Humana Press.

Page 389: Determining the Oncological and Immunological Effects of

352

A.1 Abstract

Immunocompromised mice are commonly utilized to study pancreatic cancer and other

malignancies. The ability to xenograft tumors in either subcutaneous or orthotopic

locations provides a robust model to study diverse biological features of human

malignancies. However, there is a dire need for large animal models that better

recapitulate human anatomy in terms of size and physiology. These models will be

critical for biomedical device development, surgical optimization, and drug discovery.

Here, we describe the generation and application of immunocompromised pigs lacking

RAG2 and IL2RG as a novel model for human xenograft studies. These SCID-like pigs

closely resemble NOD scid gamma mice and are receptive to human tumor tissue, cell

lines, and organoid xenografts. However, due to their immunocompromised nature,

these immunocompromised animals require housing and maintenance under germfree

conditions. In this protocol, we describe the use of these pigs in a subcutaneous tumor

injection study with human PANC1 cells. The tumors demonstrate a steady, linear

growth curve, reaching 1.0 cm within 30 days post-injection. The model described here

is focused on subcutaneous injections behind the ear. However, it is readily adaptable

for other locations and additional human cell types.

A.2 Keywords

Immunocompromised, Porcine, Cancer, Pancreatic, Ultrasound, Therapy

Page 390: Determining the Oncological and Immunological Effects of

353

A.3 Introduction

Pancreatic cancer is expected to become the second leading cause of cancer

related deaths by 2020, with patient survival rates below 9% due to late diagnosis and

poor treatment options.[1] The current armamentarium for treating pancreatic cancer is

rather limited, but still includes standard oncological care: surgery, chemotherapy,

radiation, immunotherapy, and ablation therapy. Unfortunately, the most effective

treatment option is surgical resection of the tumor, which is not a viable strategy for

patients that have advanced disease or co-morbidities.[2] The frontline chemotherapy,

gemcitabine, has been one of the most significant improvements to standard of care for

pancreatic cancer, but only improved the one-year survival from 2% to 18%.[2,3] The

scarcity of treatment options is due to late diagnosis, complicated stromal tissue, and

lack of appropriate models for studying human disease predictably in pre-clinical

studies.

In recent years, advancements in cancer therapeutics, such as minimally

invasive ablation modalities, have shown promise in pre-clinical trials and early clinical

work. For example, irreversible electroporation (IRE) ablates tumors by generating

short, high-amplitude electric fields, preferentially killing tumor cells in the ablation zone,

while leaving healthy tissue relatively intact.[4-6] Human trials have shown promise for

IRE in ablating pancreatic tumors, with patients having significant increases in overall

survival and minimal adverse events.[7-11] Focused Ultrasound therapies have also

been developed for non-invasive tumor ablation, including high intensity focused

ultrasound (HIFU) and histotripsy. HIFU has been developed as a thermal ablation

modality and has been found to improve patient outcomes in pre-clinical and clinical

Page 391: Determining the Oncological and Immunological Effects of

354

trials.[12,13]Histotripsy is a non-invasive focused ultrasound ablation technology that

uses cavitation at a focal point to ablate the targeted tissue into an acellular liquid

homogenate.[14] With the precision and tissue selectivity seen in pre-clinical trials to

date, there is hope that histotripsy will be able to overcome the negative side effects

typically reported with other more invasive ablation modalities.[15-17] However, with

IRE, histotripsy, and other emerging technologies, there are still many questions that

need to be answered about safety and efficacy that are difficult to address using

currently available animal models.

In order to better recapitulate human disease, traditional approaches have

focused on using genetically modified mouse lines.[18,19] While mice have proven to

be valuable for the study of basic cancer biology, they often fail to seamlessly translate

therapeutic strategies to human disease due to their size, differences in immune

system, lifespan, telomerase activity, genome, and other basic biological functions.

Thus, there is a need for an animal model that more predictably recapitulates human

anatomy and physiology. This need can be addressed utilizing pigs. As a model

organism pigs are similar to humans in size, genome, metabolism, telomerase activity,

and organ micro-structures.[20,21] Genetically modified pig models have included

animals with APC and TP53 mutations that developed colorectal polyps and osteogenic

tumors, respectively, similar in pathogenesis to human malignancies.[22,23] In order to

increase the breadth of tumorigenic sites, the “Oncopig” was developed with KRAS and

TP53 mutations. When stimulated by Cre recombinase activation, these pigs develop

tumor nodules at the site of Cre injection. [24,20] However, while these models have

been useful for specific studies, they are associated with significant limitations that have

Page 392: Determining the Oncological and Immunological Effects of

355

limited their use for many applications. Specifically, these models do not fully

recapitulate human disease, are not reproducible on a short time scale, and lack the

complete control of features such as growth dynamics and tumor location that is

available in well-established murine models.

In order to address this specific niche, we have generated pigs with severe

combined immunodeficiency (SCID)-like disease that can be used to reliably grow

tumors and are receptive to engraftment with human cells. These pigs are designed to

be engrafted with a diverse range of human tumor cell lines and tissues either

orthotopically or subcutaneously. The tumors can be implanted in highly controlled sites

within each tissue (i.e. in close proximity to critical structures, such as blood vessels or

ducts), and the growth curve for xenografts can be highly standardized. There are two

established means to achieve SCID-like pigs: naturally occurring or TALEN-mediated

mutations.[25,26] Both of these immunocompromised animals have been previously

established as having the ability to sustain xenograft growth.[25,26] While both SCID-

like pigs have the potential to support tumor growth, the CRISPR/Cas9 derived mutants

are the most logistically sound to utilize given the reliable derivation of the

animals.[25,27,28]

A.4 Materials

A.4.1 Generating RAG2/IL2RG Deficient Pigs

1. Humidified incubator (38.5 °C, 5% CO2, 5% O2).

2. Stereo microscope with warm plate (Nikon).

Page 393: Determining the Oncological and Immunological Effects of

356

3. Micromanipulator system (Narishige).

4. Microcapillary Puller (Shutter Instrument).

5. FemtoJet (Eppendorf).

6. Glass capillary tubes.

7. Captrol III® micropipet (Drummond Scientific).

8. 100 × 15 mm plastic petri dishes.

9. 35 × 10 mm plastic petri dishes.

10. Manipulation medium: medium 199 supplemented with 0.6 mM NaHCO3, 2.9 mM

Hepes, 30 mM NaCl, 10 ng/ml gentamicin, and 3 mg/ml BSA; pH 7.4.

11. PZM3 culture medium

12. Mineral oil, embryo tested.

A.4.2 Maintaining Cells in Culture

1. Sterile Dulbecco’s Modified Eagle’s Medium (DMEM)

2. Sterile Fetal Bovine Serum (FBS)

3. Sterile Penicillin/streptomycin (optional)

4. Sterile TC vented-cap flasks (25, 75, and 175 cm2 sizes) (see Note 1)

5. Sterile 1x Phosphate Buffered Saline (PBS)

6. 0.25% Trypsin in EDTA

7. 5, 10, 25, and 50 mL serological pipettes

8. 1000 uL pipette tips

9. Incubator (5% CO2)

Page 394: Determining the Oncological and Immunological Effects of

357

A.4.3 Harvesting Cells for Injection

1. 15 mL conical centrifuge tubes

2. 1000 uL pipette tips

3. 10 uL pipette tips

4. Microcentrifuge tube 1.7 mL

5. Refrigerated centrifuge capable of 125xg

6. Hemocytometer (see Note 2)

7. Trypan blue

8. Matrigel

A.4.4 Sterile Passing in of the Cells

1. Icepack (see Note 3)

2. Small plastic box for spraying in cells

3. Spor-Klenz

A.4.5 Injecting Tumors

1. 1 mL syringes

2. 27-gauge 1/2-inch needles

A.4.6 Monitoring Tumors and Health

1. Plastic Vernier calipers (see Note 4)

2. Rope (8-12 inches long, 1/4 inch diameter) (see Note 5)

3. Health monitoring sheets

Page 395: Determining the Oncological and Immunological Effects of

358

A.4.7 Ultrasound Imaging (see Note 6)

4. Large container

5. Electronic trimmers for hair removal

6. Laptop Computer

7. SmartUs EXT-1M beamformer (Telemed, Lithunia, EU)

8. AC power cable

9. 100~240 VAC, 50~60 Hz power supply (EN60601-1, Telemed, Lithunia, EU)

10. CTG to USB adaptor cable

11. EchoWave II Imaging software (Telemed, Lithunia, EU)

12. Linear array ultrasound imaging probe (L18-10L30H-4, Telemed, Lithunia, EU)

13. Phased array ultrasound imaging probe (P8-3L10SI-6, Telemed, Lithunia, EU)

14. Sterile water, PBS, or saline

15. Sterile gauze or paper towels

16. Ultrasound jelly

A.4.8 Necropsy

1. Scalpel or sharp, non-serrated knife

2. Gloves

3. Ruler

4. 10% formalin

5. Histology cassettes

6. Cutting board or necropsy table

7. 10% Bleach

Page 396: Determining the Oncological and Immunological Effects of

359

8. Disposable wypalls or paper towels

A.5 Methods

A.5.1 Generating RAG2/IL2RG Deficient Pigs

1. Conduct in vitro fertilization (IVF) to generate zygotes that will be injected with

CRISPR/Cas9 system targeting RAG2 and IL2RG. (see Note 7)

2. After 2-3 hours post-IVF, wash presumable zygotes in manipulation medium.

3. Place the embryos in a microinjection dish (10-20 µl drop of manipulation

medium covered with mineral oil in 100 x 10 mm petri dish).

4. Inject sgRNAs targeting RAG2 or IL2RG genes and Cas9 mRNA (10 ng/µl

sgRNA and 20 ng/µl Cas9 mRNA are recommended) into cytoplasm of the

presumable zygotes using FemtoJet microinjector under the control of

micromanipulator. Microinjection is conducted on a heating plate at 37 °C. (see

Note 8)

5. After microinjection, wash the embryos twice in PZM3 medium.[29]

6. Place the microinjected embryos in a culture dish (20 µl drops of PZM3 medium

covered with mineral oil in 35 mm dish) and incubate them at 38.5°C, 5% O2, and

5% CO2 in humidified air until the day of embryo transfer.

7. For embryo transfer, identify surrogate sows displaying signs of ovulation. (see

Note 9)

Page 397: Determining the Oncological and Immunological Effects of

360

8. Transfer the CRISPR/Cas9 injected embryos (about 50-100 embryos depending

on the developmental stage of embryos), cultured for 3-5 days, into the oviduct of

the sows through surgical embryo transfer. (see Note 10)

9. Determine pregnancy by ultrasound at day 30 of gestation.

10. Conduct a gnotobiotic hysterectomy between day 113-115 post fertilization to

derive SCID-like piglets, and then house in gnotobiotic isolators.[30] (see Note

11)

A.5.2 Maintaining Cells in Culture

1. Culture PANC1 cells in DMEM supplemented with 10% fetal bovine serum and

optional 1% penicillin/streptomycin at 37oC and 5% CO2 in tissue culture treated

flasks. (see Note 12)

2. Split cells to keep confluency between 50-80%

a. Aspirate spent media, and wash plate with room temperature PBS.

b. Cover base of the plate with 0.25% trypsin in EDTA, and incubate 5-10

minutes at 37oC and 5% CO2.

c. Add 37oC serum containing culture media, and passage desired

percentage of cells to new flask and add appropriate volume of 37oC fresh

media (i.e. 50ml for a T175).

A.5.3 Harvesting Cells for Injection

Page 398: Determining the Oncological and Immunological Effects of

361

1. Aspirate culture media, wash with PBS, cover base of the plate with 0.25%

trypsin in EDTA, and incubate 5-10 minutes at 37oC and 5% CO2. Record the

volume of trypsin used.

2. Add serum containing media to deactivate the trypsin and transfer to a 15 ml

conical tube. Record the volume added, which should be 5-8 times the amount of

trypsin used.

3. Transfer 10 uL of cell suspension to a 2mL tube, or smaller, for counting.

4. Centrifuge the 15mL tube at 125xg for 5-10 mins at 4oC.

5. Determine cell concentration using 10ul cells aliquoted with a hemocytometer.

(see Note 13)

6. Remove media from 15mL tube, and resuspend cells in Matrigel for desired cell

concentration and keep in a 1.7 ml tube. (see Notes 14-15)

7. Keep 1.7 mL tube on an ice pack.

8. Aliquot additional 1.7 tube of Matrigel, of equal volume, for control injections.

(see Note 16)

A.5.4 Sterile Passing in of the Cells

1. Place tubes of Matrigel and Matrigel-cells on ice packs in labeled plastic

containers.

2. Set plastic containers inside the gnotobiotic transport isolator port and spray

down with Spor-Klenz.

3. Seal port from transport isolator to two housing isolators with a “Y” cap. Let set

for one hour.[30] (see Note 17)

Page 399: Determining the Oncological and Immunological Effects of

362

4. Have one individual enter the transport isolator and another enter the home

isolator unit.

5. Open the port openings within both isolators.

6. Carefully, use inside arms to pass plastic container with Matrigel and Matrigel-

cells into home isolator units prior to moving any piglets. (see Note 18)

A.5.5 Injecting Tumors

Figure 0-1: Creating “Tent” to Ensure Subcutaneous Injection. By pulling the pig’s ear forward, a small tent of skin will pull up forming a small tent. To ensure a subcutaneous injection, have the bevel of the needle enter “tent” following a path indicated

1. Pull 0.3-0.6mL Matrigel (without cells) into 1mL syringe. Place 27-gauge 1/2-inch

needle on syringe and return to ice. (see Note 19)

2. Prepare second syringe of Matrigel-cells the same as above, and return to ice.

3. Have handler restrain pig by having the animal rest on one arm, while having

their free hand grasp the pig’s back for stability.

4. Pull the ear of interest away from the site of injection for control Matrigel, creating

a skin-tent (Figure 1). Carefully insert the needle bevel up into this area.

Page 400: Determining the Oncological and Immunological Effects of

363

5. Inject 100 uL when it is clear the bevel is within the skin-tent, and therefore a

subdermal injection. Pull the needle out in a straight line with a slight wrist-

twisting motion to help avoid any trailing tumor.

6. Once complete, inject the second ear on the same animal before moving on to

the next pig.

7. Make sure that neither injection leaked prior to returning pig to its compartment.

A.5.6 Monitoring Tumors and Health

Figure 0-2: Measuring Subcutaneous Tumor Growth in Gnotobiotic Porcine Isolators. (A) Subcutaneous tumors (indicated by arrow) grown behind the ear can be easily viewed within two days post injection and (B) measured with plastic Vernier calipers. (C) The

1. To measure tumors, have handler restrain the pig by having the animal rest on

one arm, while having their free hand grasp the pig’s back for stability.

2. Gently pull the pig’s ear towards their nose in order to have a clear view of the

injection site, or tumor once growth has been noted.

3. If there is palpable or viewable growth, use calipers to measure the tumor.

Determine the diameter by measuring the widest portion of the growth and the

width perpendicular to that. (Figure 2)

Page 401: Determining the Oncological and Immunological Effects of

364

4. Calculate diameter by taking the square root of the product of the two numbers

collected above.

5. Once tumor and control injection sites have been monitored, have restrainer hold

the pig by the hind legs.

6. Secure a rope with a slip knot above the pig’s ankle, by sliding the foot through

the opening in the rope and pulling the rope tight.

7. At the other end of the rope, ensure a good grip on the hanging scale.

8. Have the restrainer gently release the pig. With the pig suspended by the

hanging scale, note the weight.

9. Once the weight has been observed and the restrainer regains control of the pig,

remove the rope from the pig’s ankle by loosening the slip knot and sliding the

rope off of the pig’s foot.

A.5.7 Ultrasound Imaging

1. Set up ultrasound imaging system prior to removing pigs from isolators. (see

Note 20)

2. Remove pigs from gnotobiotic isolator by removing both port seals, and having

one person pass pigs out one at a time to another person. (see Notes 21-22)

3. Wash tumor area and shave using electronic trimmers to minimize imaging

interference. (see Note 23)

4. Apply ultrasound jelly to area, and gently press ultrasound probe onto tumor.

5. Use Telemed imaging software to collect desired image.

Page 402: Determining the Oncological and Immunological Effects of

365

A.5.8 Necropsy

1. Euthanize pigs through desired, IACUC approved methods.

2. Make a posterior cervical incision from the base of the neck to the top of the skull

to allow the skin the be folded outwards.

3. Locate the tumor within the dermis by following the visual markers from the

exterior and palpating the extent of the tumor margins within.

4. Carefully extract the mass. (see Note 24)

5. Make a midline abdominal incision from the base of the ribs down to the pelvic

bone.

6. Carefully remove the vascularized organs that are frequent sites of metastasis:

liver, spleen, and lungs.

7. One at a time preform a bread slice on each of the organs collected to check for

gross metastases.

a. Starting at one end of the organ, make sequential slices each centimeter

until reaching the opposite end of the tissue.

b. Briefly check each tissue for any gross changes in color or shape that

might be indicative of a tumor.

c. Collect any suspect tissue in a histology cassette and store in 10%

formalin.

8. Clean surface with 10% bleach and disposable Wypalls or paper towels when

done.

Page 403: Determining the Oncological and Immunological Effects of

366

A.6 Notes

1. Our preference is for TC vented-cap flasks; however, utilization of other TC

flasks or plates is based on lab preference.

2. Automatic cell counter can also be used if available.

3. Plastic, sealed ice packs are advised to be used in order to help maintain sterility

when passing cells into gnotobiotic isolators. Whether these are malleable or

solid plastic is based on availability of icepacks and plastic containers for holding

icepacks and cells together.

4. Metal Vernier calipers will corrode after being sterilized for gnotobiotic isolators.

They can be used if necessary, but it is highly advised to utilize plastic as much

as possible.

5. Exact size of rope is not critical. The important part is that the material is smooth

and will not injury the pigs during weighing, and is cut to a length that is long

enough to support two slip knots.

6. Clinical ultrasound imaging systems can be utilized based on availability and

tumor anatomical location. If feasible, using CT or MR’ imaging can also be used

for monitoring orthotopic or metastatic tumor growth.

7. Technical details of the IVF procedures can be found in a previous

publication.[31]

8. The preparation of sgRNA and Cas9 mRNA can be found in a previous

publication.[32]

9. Estrus stage of surrogate sows should be synchronized with developmental

stage of the embryos injected with CRISPR/Cas9 system.

Page 404: Determining the Oncological and Immunological Effects of

367

10. In vitro-derived pig embryos have low viability. The use of in vivo-derived

embryos may lower the number of embryos transferred to produce SCID-like

pigs.

11. While it is not critical to an oncological study to utilize gnotobiotic housing, for the

health of the SCID-like pigs, it is advised to follow gnotobiotic pig protocols for

hysterectomy delivery and housing to avoid unintending illness.

12. If utilizing an alternative cell line, use appropriate media. For example, if using a

cell line provided by ATCC, use media advised on their website.

13. If it is available, or is a common practice, use an automatic cell counter.

14. We saw successful engraftment with 1.2x106 cells in 100 uL of Matrigel, however

going as high as 6-10x107 cells should also produce viable tumors, but be aware

the increased injection density will increase the initial growth rate.

15. If you use a larger tube, or a larger syringe than advised below (3.4 step 1), it will

be difficult to administer the smaller volume for the injection.

16. Have pairs of tubes designated to go to each gnotobiotic pig isolator.

17. Once the one-hour time has passed, the port is considered sterile. If sterility is

not required (i.e. for a very short study), the incubation time can be shortened.

18. If necessary, pigs can be passed into their home isolators first, and cells passed

in at a later time. However, the pigs will be less effected by anesthesia and more

difficult to inject the further from the hysterectomy that one waits.

19. It is best to not pull too much Matrigel into a syringe at a time, because it will

make it harder to control the injection leading to distorted tumor shapes or miss

calculated injection volumes.

Page 405: Determining the Oncological and Immunological Effects of

368

20. Monitoring of tumor growth subcutaneously can be done using a linear array

ultrasound (L18-10L30H-4, TELEMED, Lithuania, EU). If you want to image

internal organs for orthotopic tumor placements or to check for metastatic

growths, utilize a phased array ultrasound probe (P8-3L0S1-6, TELEMED,

Lithuania, EU) in order to image a larger, deeper area.

21. If multiple pigs are in the isolator, have a large container to hold the pigs in until

each pig is euthanized individually. The isolator will collapse once it is opened so

you need a container to hold the pigs until they are euthanized.

22. Pass pigs out head first, and they will help by running in the correct direction, into

the arms of the catching handler.

23. To minimize risk of infection to immunocompromised animals when imaging

during study, clean area with sterilized water, PBS, or saline with sterilized

gauze, paper towels, or similar cloth.

24. When extracting the tumor, extracting extra margins can increase the probability

of removing the full tumor if full removal is the priority.

Page 406: Determining the Oncological and Immunological Effects of

369

A.7 References

1. Aier I, Semwal R, Sharma A, Varadwaj PK (2019) A systematic assessment of

statistics, risk factors, and underlying features involved in pancreatic cancer.

Cancer epidemiology 58:104-110

2. Ghaneh P, Costello E, Neoptolemos JP (2007) Biology and management of

pancreatic cancer. Gut 56 (8):1134-1152

3. Burris Hr, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR,

Christine Cripps M, Portenoy RK, Storniolo AM, Tarassoff P (1997)

Improvements in survival and clinical benefit with gemcitabine as first-line

therapy for patients with advanced pancreas cancer: a randomized trial. Journal

of clinical oncology 15 (6):2403-2413

4. Davalos RV, Mir L, Rubinsky B (2005) Tissue ablation with irreversible

electroporation. Annals of biomedical engineering 33 (2):223

5. Ivey JW, Latouche EL, Sano MB, Rossmeisl JH, Davalos RV, Verbridge SS (2015)

Targeted cellular ablation based on the morphology of malignant cells. Scientific

reports 5:17157

6. Ivey J, Wasson E, Alinezhadbalalami N, Kanitkar A, Debinski W, Sheng Z, Davalos

R, Verbridge S (2019) Characterization of Ablation Thresholds for 3D-Cultured

Patient-Derived Glioma Stem Cells in Response to High-Frequency Irreversible

Electroporation. Research 2019:8081315

Page 407: Determining the Oncological and Immunological Effects of

370

7. Martin RC, McFarland K, Ellis S, Velanovich V (2013) Irreversible electroporation in

locally advanced pancreatic cancer: potential improved overall survival. Annals of

surgical oncology 20 (3):443-449

8. Martin RC, Kwon D, Chalikonda S, Sellers M, Kotz E, Scoggins C, McMasters KM,

Watkins K (2015) Treatment of 200 locally advanced (stage III) pancreatic

adenocarcinoma patients with irreversible electroporation: safety and efficacy.

Annals of surgery 262 (3):486-494

9. Bower M, Sherwood L, Li Y, Martin R (2011) Irreversible electroporation of the

pancreas: definitive local therapy without systemic effects. Journal of surgical

oncology 104 (1):22-28

10. Martin II RC, McFarland K, Ellis S, Velanovich V (2012) Irreversible electroporation

therapy in the management of locally advanced pancreatic adenocarcinoma.

Journal of the American College of Surgeons 215 (3):361-369

11. Martin RC (2013) Irreversible electroporation of locally advanced pancreatic head

adenocarcinoma. Journal of Gastrointestinal Surgery 17 (10):1850-1856

12. Saccomandi P, Lapergola A, Longo F, Schena E, Quero G (2018) Thermal ablation

of pancreatic cancer: A systematic literature review of clinical practice and pre-

clinical studies. International Journal of Hyperthermia 35 (1):398-418

13. Hwang JH, Wang Y-N, Warren C, Upton MP, Starr F, Zhou Y, Mitchell SB (2009)

Preclinical in vivo evaluation of an extracorporeal HIFU device for ablation of

pancreatic tumors. Ultrasound in medicine & biology 35 (6):967-975

Page 408: Determining the Oncological and Immunological Effects of

371

14. Bader KB, Vlaisavljevich E, Maxwell AD (2019) For whom the bubble grows:

physical principles of bubble nucleation and dynamics in histotripsy ultrasound

therapy. Ultrasound in medicine & biology

15. Longo KC, Knott EA, Watson RF, Swietlik JF, Vlaisavljevich E, Smolock AR, Xu Z,

Cho CS, Mao L, Lee FT, Jr., Ziemlewicz TJ (2019) Robotically Assisted Sonic

Therapy (RAST) for Noninvasive Hepatic Ablation in a Porcine Model: Mitigation

of Body Wall Damage with a Modified Pulse Sequence. Cardiovasc Intervent

Radiol 42 (7):1016-1023. doi:10.1007/s00270-019-02215-8

16. Vlaisavljevich E, Owens G, Lundt J, Teofilovic D, Ives K, Duryea A, Bertolina J,

Welling TH, Xu Z (2017) Non-invasive liver ablation using histotripsy: preclinical

safety study in an in vivo porcine model. Ultrasound in medicine & biology 43

(6):1237-1251

17. Vlaisavljevich E, Kim Y, Allen S, Owens G, Pelletier S, Cain C, Ives K, Xu Z (2013)

Image-guided non-invasive ultrasound liver ablation using histotripsy: Feasibility

study in an in vivo porcine model. Ultrasound in medicine & biology 39 (8):1398-

1409

18. Wagner M, Greten FR, Weber CK, Koschnick S, Mattfeldt T, Deppert W, Kern H,

Adler G, Schmid RM (2001) A murine tumor progression model for pancreatic

cancer recapitulating the genetic alterations of the human disease. Genes &

Development 15 (3):286-293

19. Herschkowitz JI, Simin K, Weigman VJ, Mikaelian I, Usary J, Hu Z, Rasmussen KE,

Jones LP, Assefnia S, Chandrasekharan S (2007) Identification of conserved

Page 409: Determining the Oncological and Immunological Effects of

372

gene expression features between murine mammary carcinoma models and

human breast tumors. Genome biology 8 (5):R76

20. Segatto NV, Remião MH, Schachtschneider KM, Seixas FK, Schook LB, Collares T

(2017) The oncopig cancer model as a complementary tool for phenotypic drug

discovery. Frontiers in pharmacology 8:894

21. Maher B (2013) Tissue engineering: How to build a heart. Nature News 499

(7456):20

22. Flisikowska T, Merkl C, Landmann M, Eser S, Rezaei N, Cui X, Kurome M,

Zakhartchenko V, Kessler B, Wieland H (2012) A porcine model of familial

adenomatous polyposis. Gastroenterology 143 (5):1173-1175. e1177

23. Sieren JC, Meyerholz DK, Wang X-J, Davis BT, Newell JD, Hammond E, Rohret JA,

Rohret FA, Struzynski JT, Goeken JA (2014) Development and translational

imaging of a TP53 porcine tumorigenesis model. The Journal of clinical

investigation 124 (9):4052-4066

24. Schachtschneider KM, Schwind RM, Newson J, Kinachtchouk N, Rizko M,

Mendoza-Elias N, Grippo P, Principe DR, Park A, Overgaard NH, Jungersen G,

Garcia KD, Maker AV, Rund LA, Ozer H, Gaba RC, Schook LB (2017) The

Oncopig Cancer Model: An Innovative Large Animal Translational Oncology

Platform. Front Oncol 7:190. doi:10.3389/fonc.2017.00190

25. Lee K, Kwon D-N, Ezashi T, Choi Y-J, Park C, Ericsson AC, Brown AN, Samuel MS,

Park K-W, Walters EM (2014) Engraftment of human iPS cells and allogeneic

porcine cells into pigs with inactivated RAG2 and accompanying severe

Page 410: Determining the Oncological and Immunological Effects of

373

combined immunodeficiency. Proceedings of the National Academy of Sciences

111 (20):7260-7265

26. Basel MT, Balivada S, Beck AP, Kerrigan MA, Pyle MM, Dekkers JC, Wyatt CR,

Rowland RR, Anderson DE, Bossmann SH (2012) Human xenografts are not

rejected in a naturally occurring immunodeficient porcine line: a human tumor

model in pigs. BioResearch open access 1 (2):63-68

27. Lei S, Ryu J, Wen K, Twitchell E, Bui T, Ramesh A, Weiss M, Li G, Samuel H,

Clark-Deener S (2016) Increased and prolonged human norovirus infection in

RAG2/IL2RG deficient gnotobiotic pigs with severe combined immunodeficiency.

Scientific reports 6:25222

28. Kang JT, Cho B, Ryu J, Ray C, Lee EJ, Yun YJ, Ahn S, Lee J, Ji DY, Jue N, Clark-

Deener S, Lee K, Park KW (2016) Biallelic modification of IL2RG leads to severe

combined immunodeficiency in pigs. Reprod Biol Endocrinol 14 (1):74.

doi:10.1186/s12958-016-0206-5

29. Yoshioka K, Suzuki C, Tanaka A, Anas IM-K, Iwamura S (2002) Birth of piglets

derived from porcine zygotes cultured in a chemically defined medium. Biology of

reproduction 66 (1):112-119

30. Yuan L, Jobst PM, Weiss M (2017) Gnotobiotic Pigs: From Establishing Facility to

Modeling Human Infectious Diseases. In: Gnotobiotics. Elsevier, pp 349-368

31. Uh K, Lee K (2017) Use of Chemicals to Inhibit DNA Replication, Transcription, and

Protein Synthesis to Study Zygotic Genome Activation. In: Zygotic Genome

Activation. Springer, pp 191-205

Page 411: Determining the Oncological and Immunological Effects of

374

32. Ryu J, Lee K (2017) CRISPR/Cas9-mediated gene targeting during embryogenesis

in swine. In: Zygotic Genome Activation. Springer, pp 231-244