You are on page 1of 41

(eBook PDF) Translational Medicine in

CNS Drug Development, Volume 29


Visit to download the full and correct content document:
https://ebooksecure.com/download/ebook-pdf-translational-medicine-in-cns-drug-dev
elopment-volume-29/
CONTENTS vii
III. lll-Suited Disease Models 214 III. Value and Impact of ADNI 241
IV. Promising Human Genome 214 IV. ADNI-Inspired Other Initiatives 243
V. Ill-Suited Phenotypes for Drug Discovery 214 V. Potential Further Enhancements for Greater Efficiency
VI. Appropriate Phenotypes for Drug Discovery 215 and Acceleration of Drug Development 244
VII. Appropriate Data Mining for Drug Discovery 215 VI. Summary 244
VIII. Facilitating Drug Discovery Through Human References 244
Genetics 216
IX. Concluding Remarks 217
References 217
18. Regulatory Perspectives on the Use
of Biomarkers and Personalized Medicine
15. Use of Cognition to Guide Decisions About in CNS Drug Development: The FDA Viewpoint 247
the Safety and Efficacy of Drugs in Early-Phase MATHANGI GOPALAKRISHNAN AND JOGARAO V.S. GOBBURU

Clinical Trials 219 I. CNS Drug Development 247


PAUL MARUFF II. Biomarkers and Personalized Medicine in Drug
Development and Regulation 248
I. Introduction 219
III. US FDA Initiatives 252
II. Measuring Cognition to Guide Decisions About
IV. Challenges and Future Perspectives 255
the Facilitatory Effects of CNS-Active Drugs 219
V. Conclusions 256
III. Measuring Cognition to Guide Decisions About
References 257
the Deleterious Effects of CNS-Active Drugs 222
IV. Use of Cognitive Tests to Demonstrate CNS Safety 223
V. Adverse Cognitive Outcomes Can Indicate the 19. Regulatory Considerations for the Use
Presence of Occult CNS Disease 225 of Biomarkers and Personalized Medicine in CNS
VI. Characteristics of Cognitive Tests for Use in Drug Development: A European Perspective 259
Early-Phase or Experimental Medicine 226
EAMON O’LOINSIGH AND ANJANA BOSE
References 226
I. Introduction 259
16. Digital Biomarkers in Clinical Drug II. The European Regulatory Network and Approach
Development 229 to Personalized Medicine 259
AMIR KALALI, SARAH RICHERSON, EMILIA OUZUNOVA, III. CNS Diseases and Personalized Medicine 261
RYAN WESTPHAL, AND BRADLEY MILLER IV. Biomarkers, PGx, CDx, and Rare Diseases 262
V. Nongenetic BMs and Novel Methodologies,
I. Introduction 229 New Clinical Trial Designs, Patient-Reported
II. Classification of Biomarkers and Digital Biomarkers 230 Outcomes (PROs), and Digital/Wearable
III. Types of Digital Biomarkers 230 Technologies 265
IV. Wearables 230 VI. Advanced Therapy Medicinal Products and
V. Mobile Applications 232 Personalized Medicine 268
VI. Vocal Biomarkers 232 VII. Miscellaneous 269
VII. Computerized Speech and Language Analysis 232 VIII. Conclusions 269
VIII. Sensored Pill Bottles 232 References 270
IX. Wearable Biosensors and Bioelectric Tattoos 233
X. Social Media 233
XI. Gamification 233 20. Regulatory Science Objectives and Biomarker
XII. Virtual Reality 233 Qualification Through Public-Private Partnerships
XIII. Machine Learning 233 Are Critical to Delivering Innovative Treatments
XIV. Value of Digital Biomarkers 233 for CNS Diseases 277
XV. Challenges 234 
DIANE T. STEPHENSON AND STEPHEN P. ARNERIC
XVI. Collaborative Opportunities 236
XVII. Validation Pathway 236 I. Background 278
XVIII. Conclusion 237 II. Evidence That Biomarkers Increase Successful
References 237 Approvals 280
III. Gaps in Building Translational Biomarkers for CNS
17. Lessons Learned From Public Private Diseases 281
Partnerships and Consortia: The ADNI IV. Regulatory Considerations to Validate Biomarkers 285
Paradigm 239 V. Recent Regulatory Initiatives to Accelerate Biomarker
ENCHI LIU Qualification and Approvals 288
VI. Conclusions and Recommendations for the Future 291
I. Introduction 239 Acknowledgements 292
II. ADNI 240 References 292
viii CONTENTS

21. The Assessment of Cognition in 24. Translational Medicine Strategies


Translational Medicine: A Contrast Between in Alzheimer’s Disease Drug Development 349
the Approaches Used in Alzheimer’s Disease and VERONIKA LOGOVINSKY
Major Depressive Disorder 297
I. Introduction 349
JOHN E. HARRISON AND SUZANNE HENDRIX
II. Overview of Adaptive Trial Designs 350
I. A Critical Review of Cognitive Assessment in Patients III. Use of Adaptive Designs in Alzheimer’s Disease Drug
With Alzheimer’s Disease 297 Development 351
II. Best Practice Guidance for the Assessment of IV. Can Developments in Oncology Trial Designs Inform
Cognition 300 AD Trials? 354
III. Measuring Cognitive Impairments and Cognitive V. Modernization of Clinical Tools for Early AD 355
Change in Patients With Depression: A Model VI. Development of More Sensitive Clinical Tools
for AD? 303 for Early AD Performance-Based Functional
IV. Conclusions and Recommendations 306 Measures 358
References 306 VII. New Cognitive Clinical Measures in Early AD 360
VIII. Conclusion 361
22. Translational Medicine Strategies in Drug References 361
Development for Neurodevelopmental
Disorders 309 25. Experimental Medicine Models in
SIDDHARTH SRIVASTAVA, MUSTAFA SAHIN, AND LISA PROCK Generalized Anxiety Disorder and Social
Anxiety Disorder 363
I. Introduction 309
DAVID S. BALDWIN AND AYMAN ABOU-AISHA
II. Considerations in Examining Animal Model Data
Relevant to Human Clinical Trials 310 I. Experimental Medicine Approaches in Anxiety
III. Rett Syndrome 311 Disorders 363
IV. Tuberous Sclerosis Complex 316 II. Generalized Anxiety Disorder and Social Anxiety
V. Fragile X Syndrome 318 Disorder 363
VI. Challenges Associated With Transitioning to Human III. Etiology of Generalized Anxiety Disorder and Social
Clinical Trials 321 Anxiety Disorder 365
VII. Opportunities 323 IV. Carbon Dioxide Inhalation as an
VIII. Conclusions 325 Experimental Medicine Model in Anxiety
References 325 Disorders 366
V. “Low-Dose” CO2 Inhalation as an Experimental
23. Translational Medicine Strategies in Drug Medicine Model of GAD 367
Development for Mood Disorders 333 VI. Experimental Approaches to Support
ZIHANG PAN, RADU C. GROVU, AND ROGER S. MCINTYRE Treatment Development in Social Anxiety
Disorder 368
I. Introduction 333 VII. Conclusions 370
II. Cognitive Deficits in MDD 333 Acknowledgments 370
III. Anhedonia in MDD 334 References 370
IV. Inflammation and Anhedonia 336
V. Tetrahydrobiopterin in the Pathophysiology of
Anhedonia 336 26. Translational Medicine Strategies in PTSD
VI. Kynurenine in the Pathophysiology of Drug Development 375
Anhedonia 337 DAN J. STEIN, WILLIE DANIELS, AND BRIAN H. HARVEY
VII. Inflammation and Cognition 338
VIII. TNF-α in the Pathophysiology of MDD 338 I. Introduction 375
IX. Treatment Strategies Targeting Anhedonia 339 II. Phenomenology and Epidemiology 376
X. Treatment Strategies Targeting Cognitive Dysfunctions III. Translational Neuroscience of PTSD 376
in MDD 340 IV. Current Pharmacotherapy of PTSD 378
XI. Conclusion 342 V. Future Pharmacotherapies for PTSD 379
Conflict of Interest Disclosures 343 VI. Conclusion 380
References 343 References 380
CONTENTS ix
27. Unmet Medical Needs in the Treatment IV. Conclusion 414
of Depression and the Clinical Development Acknowledgments 414
References 414
of a Differentiated Antidepressant:
A Translational Line of Evidence 383
30. Hypothesizing Major Depression as a Subset
GEORGE G. NOMIKOS
of Reward Deficiency Syndrome (RDS) Linked
I. Introduction 383 to Polymorphic Reward Genes: Considerations
II. Cognitive Impairment in MDD 384 for Translational Medicine Approaches for Future
III. Vortioxetine (Trintellix): A Multimodal Drug Development 419
Differentiated Antidepressant 385
KENNETH BLUM, MARK S. GOLD, EDWARD J. MODESTINO,
IV. Vortioxetine Shows a Distinct Profile in Cognitive IGOR ELMAN, DAVID BARON, RAJENDRA D. BADGAIYAN,
Performance in Depression 385 AND ABDALLA BOWIRRAT
V. Conclusion 386
References 387 I. Introduction 419
II. Have We Hatched a Customized “Precision Medicine?” 423
28. Translating Neurobiology into Practice in III. Initial DNA Customized Studies 423
Tobacco, Alcohol, Drug, and Behavioral IV. Summary 424
References 424
Addictions 389
A. BENJAMIN SRIVASTAVA AND MARK S. GOLD
31. Traveling Through the Storm: Leveraging
I. Introduction 389 Virtual Patient Monitoring and Artificial
II. General Principles 389 Intelligence to Observe, Predict, and Affect
III. Alcohol 392 Patient Behavior in CNS Drug Development 427
IV. Opioids 394
ADAM HANINA AND LAURA SHAFNER
V. Psychostimulants: Cocaine and Amphetamines 395
VI. Marijuana 395 I. Drawing System Analysis From Weather Patterns 427
VII. Tobacco 396 II. Complexity and Noise Through Intermittent Data
VIII. NMDA Antagonists: Ketamine and PCP 396 Capture and Interpretation in Clinical Trials 428
IX. Behavioral and Process Addictions 397 III. Information Hierarchy, Standardization, and Scale 428
X. Conclusion 398 IV. Traveling With the Storm: Objective Endpoint
References 398 Assessment 429
V. No Drug Exposure, No Signal 431
29. Translational Medicine Strategies for Drug VI. Methods to Increase Drug Exposure for Greater
Development for Impulsive Aggression 403 Statistical Power: Trial Enrichment Strategies 431
EMIL F. COCCARO, ROYCE LEE, AND NEAL G. SIMON VII. Accurate Dosing Profiles 432
VIII. Predicting Patient Behavior 433
I. Cognitive/System Neuroscience and Neurobiological IX. Identifying Professional Subjects and Concealment
Substrates of Impulsive Aggressive Behavior in Humans 403 Not to Take the IMP 433
II. Testing the Antiaggressive Efficacy of X. Where Is This All Going? 433
Psychopharmacologic Agents 409 References 433
III. Experimental Medicine Approaches to the
Development of Antiaggressive Agents 411 Index 435
This page intentionally left blank
Contributors

Ayman Abou-Aisha Mood and Anxiety Disorders Service, Abdalla Bowirrat Division of Anatomy, Biochemistry and
Southern Health NHS Foundation Trust, Southampton, Genetics, Faculty of Medicine and Health Sciences, An-Najah
United Kingdom National University, Nablus, Palestine
Stephen P. Arneric Critical Path for Alzheimer’s Disease, John R. Cirrito Department of Neurology, Washington
Critical Path Institute, Tucson, AZ, United States University School of Medicine, St. Louis, MO, United States
Rajendra D. Badgaiyan Department of Psychiatry, South Emil F. Coccaro Clinical Neuroscience and
Texas Veteran Health Care System, Audie L. Murphy Psychopharmacology Research Unit, Department of
Memorial VA Hospital; Long School of Medicine, University Psychiatry, Pritzker School of Medicine, University of
of Texas Medical Center, San Antonio, TX, United States Chicago, Chicago, IL, United States
David S. Baldwin Clinical and Experimental Sciences, Patricia E. Cole Cole Imaging and Biomarker Consulting,
Faculty of Medicine, University of Southampton; Mood and LLC, Glenview, IL, United States
Anxiety Disorders Service, Southern Health NHS Foundation Joyce Colussi-Mas Victoria University of Wellington, School
Trust, Southampton, United Kingdom; University of Psychology, Behavioural Neurogenetics Group,
Department of Psychiatry and Mental Health, University of Wellington, New Zealand
Cape Town, Cape Town, South Africa
Paul Cumming Department of Nuclear Medicine, Inselspital,
David Baron Western University Health Sciences, Graduate University of Bern, Bern, Switzerland; School of Psychology
School of Biomedical Sciences, Pomona, CA; Division of and Counselling and IHBI, Queensland University of
Neuroscience and Addiction Research, Pathway Healthcare, Technology, and QIMR Berghofer Medical Research Institute,
Birmingham, AL, United States Brisbane, QLD, Australia
Nicolas Barthelemy Department of Neurology, Willie Daniels Department of Physiology, University of the
Washington University School of Medicine, St. Louis, MO, Witwatersrand, Johannesburg, South Africa
United States
Elizabeth C. de Lange Leiden Academic Centre for Drug
Randall J. Bateman Department of Neurology, Research, Gorlaeus Laboratories, Leiden, The Netherlands
Washington University School of Medicine, St. Louis, MO,
Dominique J.-F. de Quervain Transfaculty Research
United States
Platform Molecular and Cognitive Neurosciences; Psychiatric
Kenneth Blum Western University Health Sciences, University Clinics; Department of Psychology, Division of
Graduate School of Biomedical Sciences, Pomona, CA; Cognitive Neuroscience, University of Basel, Basel,
Division of Addiction Services, Dominion Diagnostics LLC, Switzerland
North Kingstown, RI; Division of Neuroscience and Aldemar Degroot Boehringer Ingelheim Pharma GmbH &
Addiction Research, Pathway Healthcare, Birmingham, AL; Co. KG., Biberach an der Riss, Germany
Division of Neuroscience Research and Addiction Therapy,
Shores Treatment and Recovery Center, Port Saint Lucie, FL; Wayne C. Drevets Janssen Research & Development, LLC, of
Human Integrated Services Unit, University of Vermont Johnson & Johnson, Titusville, NJ, United States
Centre for Clinical & Translational Science, College of Steven G. Einstein Janssen Research and Development,
Medicine, Burlington, VT; Division of Clinical Neurology, Titusville, NJ, United States
Path Foundation NY, New York, NY; Division of Addiction Bart A. Ellenbroek Victoria University of Wellington, School
Research & Therapy, Nupathways, Innsbrook, MO; Division of Psychology, Behavioural Neurogenetics Group,
of Precision Medicine, Geneus Health LLC, San Antonio, TX; Wellington, New Zealand
Department of Psychiatry, Wright State University,
Igor Elman Department of Psychiatry, Wright State
Boonshoft School of Medicine, Dayton, OH, United States;
University, Boonshoft School of Medicine, Dayton, OH,
E€otv€
os Loránd University, Institute of Psychology, Budapest,
United States
Hungary
Audrey Gabelle Memory Research and Resources Center,
James G. Bollinger Department of Neurology, Washington Gui de Chauliac Hospital, Universite de Montpellier,
University School of Medicine, St. Louis, MO, United States Montpellier, France
Peter Bonate Astellas Pharma, Northbrook, IL, United States B.M. Geiger Department of Biomedical and Nutritional
Anjana Bose Synchrogenix, A Certara Company, Delaware Sciences, Zuckerberg College of Health Sciences, University
Corporate Center, Wilmington, DE, United States of Massachusetts, Lowell, Lowell, MA, United States

xi
xii CONTRIBUTORS

Jogarao V.S. Gobburu Center for Translational Medicine, Veronika Logovinsky Regeneron Pharmaceuticals Inc.,
School of Pharmacy, University of Maryland, Baltimore, Tarrytown, NY, United States
Baltimore, MD, United States Brendan Lucey Department of Neurology, Washington
Mark S. Gold Division of Neuroscience and Addiction University School of Medicine, St. Louis, MO, United States
Research, Pathway Healthcare, Birmingham, AL; Matthew Macaluso Department of Psychiatry and
Department of Psychiatry, Washington University School of Behavioral Sciences, University of Kansas School of Medicine-
Medicine, Saint Louis, MO, United States Wichita and KU-Wichita Clinical Trials Unit, Wichita, KS,
Mathangi Gopalakrishnan Center for Translational United States
Medicine, School of Pharmacy, University of Maryland, Gerard J. Marek Astellas Pharma Global Development, Inc.,
Baltimore, Baltimore, MD, United States Development Medical Science, CNS and Pain, Northbrook,
Radu C. Grovu Mood Disorders Psychopharmacology Unit IL, United States
(MDPU), Toronto Western Hospital, University Health Paul Maruff Florey Institute for Neuroscience and Mental
Network, Toronto, ON, Canada Health, University of Melbourne, Parkville, VIC, Australia;
Gerhard Gr€under Department of Molecular Neuroimaging, Cogstate, New Haven, CT, United States
Central Institute of Mental Health (CIMH), Mannheim, Roger S. McIntyre Mood Disorders Psychopharmacology
Germany Unit (MDPU), Toronto Western Hospital, University Health
Adam Hanina AiCure, New York, NY, United States Network; Institute of Medical Science; Department of
John E. Harrison Metis Cognition Ltd., Kilmington Common, Pharmacology; Department of Psychiatry, University of
Kilmington; Institute of Psychiatry, Psychology and Toronto; Brain and Cognition Discovery Foundation,
Neuroscience, King’s College London, London, United Toronto, ON, Canada
Kingdom; Alzheimer’s Center, VUmc, Amsterdam, The Bradley Miller Eli Lilly and Company, Indianapolis, IN,
Netherlands United States
Brian H. Harvey Division of Pharmacology, School of Timothy Miller Department of Neurology, Washington
Pharmacy and Centre of Excellence for Pharmaceutical University School of Medicine, St. Louis, MO, United States
Sciences, North-West University, Potchefstroom, South Edward J. Modestino Department of Psychology, Curry
Africa College, Milton, MA, United States
Suzanne Hendrix Pentara Corporation, Salt Lake City, UT, George G. Nomikos Biogen, Clinical Development,
United States Cambridge, MA, United States
Christophe Hirtz Laboratoire de Biochimie Proteomique Eamon O’Loinsigh Synchrogenix, A Certara Company,
Clinique, CHU de Montpellier, Universite de Montpellier, Delaware Corporate Center, Wilmington, DE, United States
INSERM U1183, Montpellier, France
Emilia Ouzunova Neuroscience Center of Excellence, IQVIA,
Yash B. Joshi Department of Psychiatry, University of Durham, NC, United States
California, San Diego, CA, United States
Zihang Pan Mood Disorders Psychopharmacology Unit
Amir Kalali San Diego, CA, United States (MDPU), Toronto Western Hospital, University Health
Meyrick Kidwell Victoria University of Wellington, School Network; Institute of Medical Science, University of Toronto,
of Psychology, Behavioural Neurogenetics Group, Toronto, ON, Canada
Wellington, New Zealand Andreas Papassotiropoulos Transfaculty Research Platform
Bruce J. Kinon Lundbeck North America, Deerfield, IL, Molecular and Cognitive Neurosciences; Department of
United States Psychology, Division of Molecular Neuroscience; Psychiatric
Paul Kotzbauer Department of Neurology, Washington University Clinics; Department Biozentrum, Life Sciences
University School of Medicine, St. Louis, MO, United States Training Facility, University of Basel, Basel, Switzerland
Michael Krams Janssen Research & Development, LLC, of Bruce W. Patterson Department of Internal Medicine,
Johnson & Johnson, Titusville, NJ, United States Washington University School of Medicine, St. Louis, MO,
United States
Royce Lee Clinical Neuroscience and Psychopharmacology
Jeffrey Paul Drexel University, Graduate School of
Research Unit, Department of Psychiatry, Pritzker School of
Biomedical Sciences, College of Medicine, Philadelphia, PA,
Medicine, University of Chicago, Chicago, IL, United States
United States
Sylvain Lehmann Laboratoire de Biochimie Proteomique
Katrina Paumier Department of Neurology, Washington
Clinique, CHU de Montpellier, Universite de Montpellier,
University School of Medicine, St. Louis, MO, United States
INSERM U1183, Montpellier, France
E.N. Pothos Program in Pharmacology and Experimental
Gregory A. Light Department of Psychiatry, University of Therapeutics and Pharmacology and Drug Development,
California, San Diego, CA, United States Sackler School of Graduate Biomedical Sciences and
Enchi Liu E-Scape Bio, South San Francisco, CA, United Department of Immunology, Tufts University School of
States Medicine, Boston, MA, United States
CONTRIBUTORS xiii
Sheldon H. Preskorn Department of Psychiatry and Center/New York State Psychiatric Institute, New York, NY,
Behavioral Sciences, University of Kansas School of Medicine- United States
Wichita and KU-Wichita Clinical Trials Unit, Wichita, KS, Siddharth Srivastava Department of Neurology, Boston
United States Children’s Hospital, Harvard Medical School, Boston, MA,
Lisa Prock Translational Neuroscience Center; United States
Developmental Medicine Center, Division of Developmental Dan J. Stein Department of Psychiatry, University of Cape
Medicine, Department of Medicine, Boston Children’s Town, Cape Town, South Africa
Hospital, Harvard Medical School, Boston, MA, United States
Diane T. Stephenson Critical Path for Parkinson’s, Critical
Sarah Richerson Neuroscience Center of Excellence, IQVIA,
Path Institute, Tucson, AZ, United States
Durham, NC, United States
Melissa A. Tarasenko Department of Psychiatry, University
Mustafa Sahin Department of Neurology; Translational
of California, San Diego, CA, United States
Neuroscience Center, Boston Children’s Hospital, Harvard
Medical School, Boston, MA, United States Joop van Gerven Centre for Human Drug Research, Leiden,
Chihiro Sato Department of Neurology, Washington The Netherlands
University School of Medicine, St. Louis, MO, United States Tim West C2N Diagnostics, LLC, St. Louis, MO, United
Jonathan Savitz Laureate Institute for Brain Research, Tulsa, States
OK, United States Ryan Westphal Eli Lilly and Company, Indianapolis, IN,
Laura Shafner AiCure, New York, NY, United States United States
Neal G. Simon Department of Biological Sciences, Lehigh Kevin Yarasheski C2N Diagnostics, LLC, St. Louis, MO,
University, Bethlehem, PA, United States United States
A. Benjamin Srivastava Department of Psychiatry, Jiun Youn Victoria University of Wellington, School of
Washington University School of Medicine, Saint Louis, MO; Psychology, Behavioural Neurogenetics Group, Wellington,
Department of Psychiatry, Columbia University Medical New Zealand
This page intentionally left blank
Preface

Development of new drugs for the treatment of CNS examples of (i) biomarker modalities and experimental
disorders has been hampered by inefficiencies in advanc- medicine in early phases of clinical development;
ing compounds from nonclinical discovery to the clinic. (ii) the uptake of these approaches by public-private part-
This may be due to the lack of appropriate mechanisms nerships, consortia, and regulators and lessons learned
for testing the pharmacology of compounds as they tran- from such interactions; and (iii) the use of translational
sition from bench to bedside. Until recently, emphasis medicine tools in clinical development efforts in selected
was placed on testing the new molecules in various indications and therapeutic areas.
in vitro and in vivo models and then promoting the best The first chapter provides examples of translation of
candidates for further evaluation in man. As a result the nonclinical models of disease and drug action to the clinic
number of approvals of new medications has remained (Geiger and Pothos), while the second chapter covers the
relatively low, while the unmet medical needs in the role of biomarkers in defining early patient studies and
treatment of CNS disorders remain high. Over the last clinical trial efficiency (Degroot). The next two chapters
decade, game-changing strides have been made in our discuss the applications of modeling and simulation in
understanding of the pathophysiology of CNS disorders the translational pharmacology of CNS drugs (Bonate
and the relation of drug exposure in plasma and CNS to and de Lange) and functional measurements of CNS drug
pharmacodynamic measures in both animals and effects in early human drug development (van Gerven).
humans. In addition, considerable progress has been We continue with a chapter on experimental medicine
made in the use of neuroimaging, neurochemical, neuro- approaches in CNS drug development (Paul), a chapter
physiological, and pharmacogenetic/pharmacogenomic on Phase I trials from traditional to newer approaches
biomarker tools in monitoring disease and drug (Macaluso et al.), a chapter on translational and experi-
responses. Thus biomarkers that play multiple roles in mental medicine approaches for antidepressant drug
the diagnosis and progression of the disease and as tools development (Marek), and a chapter on biomarker
in showing central exposure, target engagement, func- opportunities for enrichment of clinical trial populations
tional responses, and patient enrichment in clinical trials for drug development in schizophrenia and depression
have been developed with a well-defined context of use. (Kinon). The next few chapters focus on describing vari-
Experimental medicine trials in healthy volunteers or ous biomarker modalities in CNS drug development;
patients also provide confidence in advancing drug these include applications of translational neuroimaging
development to exploratory clinical trials. The utility of research (Cole and Einstein), PET occupancy and compe-
these approaches will ultimately be tested, as drugs pro- tition in translational medicine (Cumming and Gr€ under),
gress successfully to confirmatory clinical trials, registra- uses of stable isotope labeling kinetics in CNS transla-
tion, and launch. Furthermore, biomarker-driven tional medicine (Bateman et al.), applications of neuro-
personalized medicine offers the opportunity to treat physiological biomarkers with focus on psychoses
patients based on their individual characteristics and (Joshi et al.), heart rate variability as a translational bio-
achieve better efficacy and safety outcomes. Given this marker for emotional and cognitive deficits (Ellenbroek
background information, it appears timely to attempt et al.), applications of pharmacogenetic and pharmacoge-
at capturing in a systematic manner the latest develop- nomic biomarkers (Papassotiropoulos and De Quervain),
ments in translational medicine and the biomarker tool the use of cognition to guide decisions about the safety
bag in use for the development of new drugs for the treat- and efficacy of drugs in early phase clinical trials
ment of CNS disorders. This volume is intended for clini- (Maruff ), and digital health-based biomarkers for use
cians; drug developers in industry and academia; and in clinical drug development (Kalali et al.). These are fol-
graduate students in behavioral neuroscience, neurobiol- lowed by chapters on lessons learned from public-private
ogy, translational neuroscience, clinical psychiatry, and partnerships and consortia based on the ADNI paradigm
pharmaceutical science. The chapters in this volume (Liu), regulatory perspectives on the use of biomarkers
cover several aspects of applications of translational med- and personalized medicine from the FDA and EMA
icine in CNS drug development by providing specific viewpoint (Gopalakrishnan and Gobburu; O’Loinsigh

xv
xvi PREFACE

and Bose), and learnings from the Critical Path Institute in tobacco, alcohol, drug, and behavioral addictions
efforts in regulatory qualification of biomarkers (Srivastava and Gold): for impulse aggression (Coccaro
(Stephenson and Arneric). The next chapter addresses et al.): and for hypothesizing major depression as a subset
the assessment of cognition in translational medicine of reward deficiency syndrome linked to polymorphic
and the contrast between the approaches used in reward genes (Blum et al.). The final chapter presents
Alzheimer’s disease and major depressive disorder practical applications of virtual patient monitoring for
(Harrison and Hendrix) followed by a number of chap- increased signal detection in CNS drug development
ters on translational medicine strategies and experimen- (Hanina and Shafner).
tal medicine approaches in drug development for We thank all the contributors of the chapters in this
neurodevelopmental disorders (Sahin and Srivastava); volume for sharing their experience and knowledge with
for mood disorders with focus on anhedonia and cogni- us and our readers, who we hope that they find the
tive dysfunction and the role of inflammatory processes present data and information useful and applicable to
(Pan et al.); in Alzheimer’s disease (Logovinsky); in gen- their translational medicine approaches in CNS drug
eralized anxiety and social anxiety disorders (Baldwin development.
and Ayman); in PTSD (Stein et al.); in the clinical devel-
opment of a differentiated antidepressant (Nomikos); George Nomikos and Douglas Feltner
C H A P T E R

1
Translating Animal Models of Obesity
and Diabetes to the Clinic
B.M. Geiger*, E.N. Pothos†
*Department of Biomedical and Nutritional Sciences, Zuckerberg College of Health Sciences, University of Massachusetts,
Lowell, Lowell, MA, United States †Program in Pharmacology and Experimental Therapeutics and Pharmacology
and Drug Development, Sackler School of Graduate Biomedical Sciences and Department of Immunology,
Tufts University School of Medicine, Boston, MA, United States

I INTRODUCTION A Pathophysiological Similarities to Human


Systems
Preclinical models of disease, specifically animal
models, are a necessary tool for a robust and innovative When considering which animal models to use, one of
drug development process. The use of animal models to the first criteria to consider should be how closely does
identify and validate pharmacological targets has been the onset and development of the disease in the animal
part of drug development for decades. Experiments in mimic the corresponding human disease. For example,
cell culture systems can provide valuable molecular mech- in humans, an increase in an individual’s body weight
anistic information, but they cannot typically model the and excess blood glucose over time will contribute to
more complex interactions of multiple cell types that often decreased sensitivity of their insulin receptors and the
contribute to the pathophysiology of a disease. Animal eventual development of type 2 diabetes (Reinehr, 2013;
models that contribute to our understanding of the CNS Weiss et al., 2004). Therefore, a good model of this disease
are especially important, as acquiring tissue to study from would also develop insulin resistance over time as body
the human brain is not generally feasible or permissible. In weight, adiposity, and blood glucose levels increase.
this chapter, we discuss the various attributes of valid ani- As an example, C57Bl/6 mouse models of diet-induced
mal models and illustrate specific examples of animals that obesity have been shown to follow the same metabolic
we use for translational research in obesity and diabetes. progression of the disease as seen in humans and develop
hyperglycemia and insulin resistance over time, simi-
larly to the human disease (Collins, Martin, Surwit, &
Robidoux, 2004; Petro et al., 2004; Speakman, Hambly,
II ATTRIBUTES OF VALID Mitchell, & Krol, 2007; Wang & Liao, 2012). In studying
ANIMAL MODELS these models, researchers have been able to elucidate
molecular mechanisms involved in the development of
Animal models can be extremely valuable in the trans- insulin resistance over time. We discuss the utility
lation of our understanding of various diseases to treat- of diet-induced obesity models in greater detail later in
ments for those diseases in the clinic. Good disease this chapter.
models have some, if not all, of the following attributes:
(1) pathophysiological similarities to human disease, (2)
B Phenotypic Match to Disease State
a phenotypic match to the disease state, (3) replicability
and/or reproducibility, and (4) cost-efficiency (Blass, Other times, models may be used, where the exact
2015). The first two criteria directly address the validity pathophysiology of a disease is not a match to the
of the model (i.e., whether and how it relates to the dis- human pathophysiology, but the presentation of the dis-
ease it is supposed to model). ease in the model is similar to the human condition.

Translational Medicine in CNS Drug Development, Volume 29 1 © 2019 Elsevier B.V. All rights reserved.
ISSN: 1569-7339
https://doi.org/10.1016/B978-0-12-803161-2.00001-1
2 1. TRANSLATING ANIMAL MODELS OF OBESITY AND DIABETES TO THE CLINIC

These models are referred to as phenotypic models of preceding text, if a group is reproducing the results from
disease. The phenotype of a model is the set of character- an MPTP-induced Parkinson’s disease model, but does
istics, usually disease-related, that can be observed. If a not have the coordinates or dosage of MPTP used in the
model is a phenotypic match to a disease, then it has sim- original model, reproduction of the original results will
ilar observable traits as a human who has that disease. be extremely difficult, as different parameters could easily
For example, the ob/ob mouse exhibits extreme weight result in ablation of a different set of neurons than the orig-
gain in the form of excess adipose tissue, similarly to inal research. Additionally, many times a study will limit
human obesity. However, the development of obesity the model to a specific gender. If the reproducing group
in the ob/ob mouse arises from leptin deficiency, while uses the opposite gender, they may find that gender differ-
the vast majority of obese individuals have high levels ences complicate their ability to reproduce the original
of leptin. Only a very small number of human obese results. Furthermore, with all animal models, reproduc-
patients are actually leptin-deficient (Farooqi, 2008; ibility is difficult when different litters of animals are
Farooqi et al., 2007; Farooqi & O’Rahilly, 2008; Wang, used. This is particularly problematic if the animals are
Chandrasekera, & Pippin, 2014). Therefore, this model from different suppliers as this introduces more variabil-
matches the phenotype of obesity, while the pathophys- ity in the animals’ genetic background and microbiome,
iology presents differences. Investigators should use which could influence the study results (Masca et al.,
caution when using models that are phenotypic models 2015). Sometimes, animals of the same species, gender,
of disease to elucidate the molecular mechanisms of the and phenotype may manifest important differences in
disease. The altered underlying physiology may not behavior, physiology, disease state, or microbiome even
match the human pathophysiology of the disease. if they come from different batches of the same or differ-
ent suppliers housed in different locations and fed differ-
ent diets (Ussar et al., 2015).
C Replicability and Reproducibility
Replicability of a model is the ability to use the same D Cost Efficiency
model within a research group and achieve similar results.
Finally, the cost of husbandry of the various animal
When conducting research using animal models, a study
models varies widely and should be considered when
will often span several months of data collection and use
determining which animal model is best suited for a par-
animals that have been born to different dams or in dif-
ticular project. The physiology of nonhuman primates is
ferent litters. In this case, replicability of the model is
very similar to human physiology. However, the regula-
extremely important. If the model is developed using
tions pertinent to a nonhuman primate facility are much
an environmental insult, like 1-methyl-4-phenyl-1,2,3,6-
more stringent than with other research models, and
tetrahydropyridine (MPTP) injections for the generation
therefore the cost associated with the establishment,
of a Parkinson’s disease rodent model, consistent use of
housing and maintenance of these facilities may not be
the same stereotaxic coordinates is necessary to ensure
feasible for many researchers. On the other hand, zebra
that the insult is in the same portion of the brain. The
fish are being used more and more as preclinical models
results will also replicate better if the same lot and batch
of disease because the construction and maintenance of
of MPTP is used throughout the duration of the study.
the facilities to house them are much less expensive
Numerous other details of the experiment must also be
(Kim, Carlson, Zafreen, Rajpurohit, & Jagadeeswaran,
controlled in order to produce results that are replicable
2009; Paige, Hill, Canterbury, Sweitzer, & Romero-
( Jackson-Lewis & Przedborski, 2007; Meredith &
Sandoval, 2014). The cost of development, housing, and
Rademacher, 2011). In the case of transgenic animal
maintenance of rodent models of disease is somewhere
models, the best design to ensure replicability is to use
in between the cost of a nonhuman primate colony and
the wild-type littermates of the knockout animals, sup-
a zebra fish facility. For most research organizations, both
plemented by the appropriate backcrossing to ensure
academic and industrial, protocols and resources for con-
genes neighboring to the targeted ones are not affected,
ducting research in rodents are well established.
as opposed to a more general wild-type animal like the
C57Bl6 mouse (Masca et al., 2015; Wolfer, Crusio, &
Lipp, 2002).
Reproducibility is the ability of other researchers to III TYPES OF ANIMAL MODELS USED
reproduce the model in their lab and achieve similar FOR TRANSLATIONAL RESEARCH
results to those of the original research. Investigators can
most easily reproduce results when the original descrip- While the vast majority of animal models are rodents,
tion of the development and use of the model is clear many different animal species are used for translational
(Masca et al., 2015). Using the same examples as in the research in drug discovery and development, ranging
III TYPES OF ANIMAL MODELS USED FOR TRANSLATIONAL RESEARCH 3
from flies and worms to monkeys and other large ani- larval fish, and other brain areas are not as developed
mals. In some cases, the species is chosen based on its in fish as they are in mammals making it difficult to
relative simplicity, while other times, it may be chosen extend the results of the study in zebra fish to mammals
based on the similarity of a particular organ system to (Kalueff, Echevarria, et al., 2014; Kalueff, Stewart, et al.,
the human system. In addition to different animal species 2014; Stewart et al., 2014). Despite these difficulties, use-
that may be used, recent advances in genetics technology ful zebra fish models related to energy balance and obe-
now allow us better control of DNA modifications in live sity have been developed. We have shown that intestinal
animal models. In this section, we discuss some of the melanin-concentrating hormone, which is primarily
more common animal models and the inducible knockout known for its appetite regulating effects, is upregulated
and CRISPR-Cas9 DNA editing technologies that are used in zebra fish with TNBS-induced enterocolitis constitut-
in research, including CNS drug development. ing a striking molecular similarity to the mouse and
human form of the disease (Geiger et al., 2013). A diet-
induced obesity model in zebra fish has also been
A Rodent Models described, which will be useful in understanding more
of the pathophysiology associated with the development
The most common species of animal model used for
of obesity (Oka et al., 2010).
translational research in the CNS is the rodent model,
usually rats or mice. These models have several advan-
tages over other types of models. First, these models
are mammalian and have similar physiology to humans
C Other Small Animal Models
for many organ systems, including the brain. While not Besides rodents, other small mammalian models may
the least expensive option for this type of research, they be used in research. These species are typically used very
are usually more affordable than the other mammalian sparingly, but there are instances when they are neces-
models that may be used. Finally, researchers are often sary due to their similarities to specific human organ sys-
interested in understanding how learning and memory tems. These species could include dogs, cats, pigs, gerbils,
are affected when developing CNS-related drugs. Robust rabbits, and hamsters. For example, if a study protocol is
experimental protocols have been developed to assess attempting to address administration of a transdermal
changes in these areas in rats and mice that can then be nociceptive agent, a pig model may be used as the skin
translated to the development of new therapeutics for of the pig has striking similarities to human skin and
diseases like Alzheimer’s or Parkinson’s disease. In a later delivery of the agent through the skin is an important
section of the chapter, we apply these principles and dis- consideration in the development of transdermal formu-
cuss the mouse and rat models used for the study of obe- lations. Cats have been found to be particularly useful in
sity in more detail. We also provide examples of how the the study of Alzheimer’s disease as they exhibit disease
information from these models can then be translated to progression very similar to that in humans (Chambers
the clinic. et al., 2015), and dogs are particularly useful in kidney
and renal disease research (Norrdin, 1981).

B Zebrafish
The use of zebra fish as a model for various diseases,
D Nonhuman Primate Models
particularly in the CNS, is becoming more common Nonhuman primates are expensive animal models that
(Kalueff, Echevarria, & Stewart, 2014; Kalueff, Stewart, & still have great utility not only in drug pharmacokinetic
Gerlai, 2014; Stewart, Braubach, Spitsbergen, Gerlai, & analysis but also in the drug discovery and development
Kalueff, 2014). In addition to the cost-effectiveness of zebra process due to their similarities to human physiology,
fish, they can be useful for many other reasons. Many of the particularly in the CNS. Their high level of intellectual
same cell types, tissues, and organ systems are similar in ability allows researchers to ask and answer questions
the zebra fish and humans, including complex systems related to complex social and cognitive behaviors for
like the stress endocrine axis. Additionally, when study- which other types of animal models cannot be used. For
ing developmental disorders, the transparency of the example, the social interactions that surround food intake
zebra fish larva allows monitoring and modification of in humans are extremely complex and difficult to study in
organ systems as they develop in vivo. However, like the uncontrolled setting of normal everyday life. How-
with any model, there are limitations to the use of zebra ever, nonhuman primates, including the macaque mon-
fish. In adult zebra fish, drug treatment is often done by key, exhibit eating behaviors that are extremely similar
solubilizing the drug in tank water, but many drugs are to humans. Researchers are able to watch and learn from
not highly water-soluble. With CNS-specific studies, primates in a more controlled social environment than
some behaviors are learned and cannot be studied in the complex set of parameters under which humans
4 1. TRANSLATING ANIMAL MODELS OF OBESITY AND DIABETES TO THE CLINIC

operate. Furthermore, many of the same gut and neuro- IV EXAMPLES OF ANIMALS MODELS
peptide systems control feeding in different situations USED FOR TRANSLATIONAL
in these animals as they do in humans, so we can place OBESITY RESEARCH
them in a specific context and evaluate how the eating
behavior is affected by the circumstances and which Because the obesity phenotype is easily identifiable and
hormones are altered. These correlations can then be quantifiable, unlike the phenotype of diseases like schizo-
used to identify and test specific drug molecules before phrenia, autism, and clinical depression, researchers
they are tested in humans (Wilson, Moore, Ethun, & involved in the discovery and development of obesity
Johnson, 2014). therapeutics have used numerous valid and reliable
animal models that focus on the CNS pathophysiology
of food intake, energy expenditure, and food reward in
the translation of disease targets to the clinic. The follow-
E Targeted Genetic Modifications of Animal
ing section gives an overview of some of these obesity-
Models
related models as examples of the use of preclinical
Some of the more recent advances in genetic modifi- models in CNS translational efforts.
cation have allowed researchers to alter the DNA of tra-
ditional animal models with unprecedented precision. A Behavioral and Environmental Models
Two of these technologies, inducible knockouts using
Cre-LoxP systems and DNA editing using CRISPR- When some of the contributing factors to the pathogen-
Cas9, are discussed in the succeeding text. esis of a disease are environmental and/or behavioral, the
preclinical models that best represent the onset and path-
ophysiology of that disease will often include a simula-
1 Inducible Knockout Animal Models tion of that environment. The rapid increase in obesity
Traditional knockout animal models eliminate the in developed countries around the world is in part due
functioning gene from the DNA of the organism at the to the environment in which we live with increased
embryo stage. This approach leads to the elimination availability of highly palatable foods and a more seden-
of the gene throughout the whole organism for the tary lifestyle. As a result, the most commonly prescribed
duration of their life and has been possible to a large treatments for obesity are diet and exercise, and some of
extent only in mice. Over the last two decades, new tech- the most important preclinical models of obesity incorpo-
nology has emerged that can alter gene expression in rate diet and/or exercise to investigate the mechanisms
both time- and tissue-specific manner through the use by which these factors contribute to the obesity pheno-
of Cre recombinase fused to a tissue-specific gene and type. In this section, we discuss models that are developed
the use of LoxP of the target gene for deletion (Gierut, based on behavioral attributes of the animal and specific
Jacks, & Haigis, 2014; Lakso et al., 1992; Lobe & Nagy, environmental cues, including diet and exercise. We also
1998). The Cre-LoxP system was used to produce a include a discussion of the use of changes in the environ-
tamoxifen-induced ablation of G protein-coupled recep- ment within the gut via the gut microbiota and its role in
tor kinase 2 (GRK2) in mice with diet-induced obesity. translational obesity research.
The use of this model validated GRK2 as a potential
drug target for the treatment of obesity (Vila-Bedmar 1 Animal Models of Dietary Obesity
et al., 2015). One particularly useful model in obesity research is the
diet-induced obesity (DIO) model as it mimics the most
common cause of obesity in humans, dietary obesity. In
2 CRISPR-Cas9 DNA Editing this model, animals, usually rats and mice, although zebra
One of the most recent major advances in model devel- fish DIO models have also been developed, are exposed to
opment was the advent of the CRISPR-Cas9 gene-editing a specific diet that has macronutrient and/or caloric differ-
technology that can precisely target and edit specific ences than their normal chow diet. There are a variety of
sequences in genes. By introducing a mutation to study types of diets that are used that range from high-sucrose
or correct a genetic deficit, this method allows for the study and high-fat diets, high-fat only diets, high-saturated fat
of this editing as a therapy (Barrangou et al., 2007; Cong only diets, to food variety (cafeteria-style) diets (Geiger
et al., 2013; Savell & Day, 2017). While research related et al., 2009; Hariri, Gougeon, & Thibault, 2010; Hariri &
to the CRISPR-Cas9 technology is still relatively new, this Thibault, 2010; Hryhorczuk et al., 2016; Oka et al., 2010).
technology provides a unique opportunity for investiga- Other variations of diets may also be used depending on
tors to further identify the effects of genetic manipula- the research question being addressed. All of these diets
tions on disease phenotypes and genotypes that have are effective at increasing body weight more rapidly in
not been previously possible in several species. the animals exposed to the diets than control animals in
IV EXAMPLES OF ANIMALS MODELS USED FOR TRANSLATIONAL OBESITY RESEARCH 5
the experiment that eat a normal lab chow diet. As a result, linked to the mesolimbic dopamine-releasing ability
you can monitor how the increasing adiposity is affecting of palatable high-energy food.
the various neurochemical pathways that are associated The first evidence of linking mesolimbic dopamine
with food intake and energy expenditure. For example, release to dietary obesity came through our investiga-
animals that ate a cafeteria-style diet increased weight tion of chronic exposure (15 weeks) of rats to a high-
very rapidly and had excess weight gain over the control energy, palatable cafeteria diet along the diet model
animals as early as 2 weeks after the start of the cafeteria developed by Anthony Sclafani (Pothos, Sulzer, &
diet (Geiger et al., 2009). These animals also showed a pref- Hoebel, 1998). We found that Sprague-Dawley rats took
erence for a sweetened condensed milk and high-sucrose the majority of their daily caloric intake from high-
solutions over other choices that included regular lab carbohydrate sources and developed diet-induced obe-
chow, chocolate, peanut butter, salami, marshmallow, sity (Geiger et al., 2009; Pothos et al., 1998). As shown
cookies, and bananas. in Fig. 1.1, DIO rats demonstrated depressed basal
One major development in obesity preclinical research dopamine release in the nucleus accumbens and an atten-
is the accumulating evidence on the role of central synap- uated dopamine response to a standard chow meal or sys-
tic plasticity on food intake and weight gain. Since the temic administration of d-amphetamine, a drug that
discovery that lesions in the hypothalamus induce obesity releases the neurotransmitter dopamine into the extracellu-
(Stricker, 2012; Teitelbaum & Stellar, 1954), there have lar space by being a substrate of the plasma membrane and
been numerous studies that focus on the CNS as the vesicular transporters for dopamine (DAT and VMAT2,
regulator of homeostatic mechanisms of energy balance respectively) (Pothos & Sulzer, 1998; Sulzer & Rayport,
through the activation of serotonin receptors and orexi- 1990). In further studies on strains of rodents that were
genic and anorexigenic circuits in the arcuate nucleus selectively bred to develop dietary obesity, we established
and through the melanocortin-4 (MC4) receptor in the that the attenuation of basal and stimulated dopamine
paraventricular nucleus of the hypothalamus (PVH) release in the brain characterized these animal models since
(Garfield & Heisler, 2009). DIO animal models have been early postnatal life (Fig. 1.2) (Geiger et al., 2008).
extensively employed in this line of research. However, Our findings of decreased dopamine availability in DIO
the rapid rise of common dietary obesity in industrialized rats were replicated in other studies with obesity animal
societies indicates that nonhomeostatic signaling path- models (Fulton et al., 2006; Geiger et al., 2008; Geiger
ways that allow for chronic positive energy intake may et al., 2009; Hryhorczuk et al., 2016; Pothos et al., 1998;
be responsible. A crucial question is why laboratory ani- Wang et al., 2001) and imaging studies in obese humans
mals and humans keep on eating energy-rich, palatable (Stice, Spoor, Bohon, & Small, 2008; Stice, Spoor, Bohon,
food to the degree that they become obese. From an evo- Veldhuizen, & Small, 2008; Wang et al., 2001) and led to
lutionary perspective, it is to be expected that the brain the theory of dopamine deficiency in obesity that
developed a system to respond to natural rewards, such expanded to the reward deficiency hypothesis. It postu-
as food. These central mechanisms are conserved across lates that hyperphagia, or hedonic food intake in excess
species in order to ensure survival (Kelley & Berridge, of energy needs, is a drive to compensate for the compro-
2002) and could interact with or modulate the circuitry mised dopamine neurotransmission in the brain of obese
that regulates body weight. Therefore, availability of animals and patients and their blunting of food reward
rewarding palatable food may lead to increased caloric (Allen et al., 2012).
intake and weight gain that homeostasis-driven mecha- Proof of principle for the importance of reversing dopa-
nisms, originating primarily in the hypothalamus, may mine deficits in the brain in order to reduce hyperphagia
not overcome. This possibility may explain, at least in is the efficacy of the dopamine stimulant D-amphetamine
part, the epidemic proportions of dietary obesity. in inducing weight loss (Harris, Ivy, & Searle, 1947).
Prominent among neural systems known to code Amphetamine was manufactured as an ephedrine ana-
for the perception of reward are the mesolimbic dopa- log in the late 19th century and used as over-the-counter
mine pathways, where the release of the neurotransmit- medication until the early 1970s for weight loss. Its with-
ter dopamine, particularly in the nucleus accumbens drawal from the market following its Schedule II classi-
and medial prefrontal cortex terminals originating fication ended the tenure of amphetamine as weight-loss
in the ventral tegmental area (VTA), along with the dor- medication and highlighted concerns over dopamine-
sal striatum (that receives dopaminergic projections releasing drugs, namely, their addictive and neurotoxic
from the substantia nigra pars compacta), is known to potential that can result in severe extrapyramidal symp-
mediate reward. The activation of these pathways toms and, in some cases, suicidality, psychosis, and other
includes elevation of dopamine levels and changes in adverse psychoactive symptoms (Power et al., 2014).
dopamine turnover after natural rewarding behaviors The case of amphetamine further strengthens the state-
like feeding (Hernandez & Hoebel, 1988). It is, there- ment that stimulation of dopamine release in the brain
fore, reasonable to expect that dietary obesity may be results in inhibition of feeding. Partial dopaminergic
6 1. TRANSLATING ANIMAL MODELS OF OBESITY AND DIABETES TO THE CLINIC

FIG. 1.1 Basal, amphetamine- and laboratory chow meal-challenged nucleus accumbens dopamine levels are decreased in freely moving dietary
obese rats in vivo. (A) Body weight of cafeteria DIO rats during a 14-week period was significantly higher than that of the laboratory chow-fed group
beginning at week 2 of the dietary regimen (*P < 0.01 by one-way ANOVA). (B) Basal and amphetamine-challenged extracellular dopamine levels
during week 14 in the nucleus accumbens of cafeteria DIO rats (n ¼ 9) was significantly lower than in chow-fed rats (n ¼ 13). (C) The percent increase
from baseline after amphetamine was higher in the cafeteria DIO rats than in the chow-fed rats (*P < 0.01 between groups, P < 0.05 within both
groups, #P < 0.05 within laboratory chow-fed group only, and ##P < 0.05 within DIO group only relative to baseline prior to the amphetamine injec-
tion). (D) A plain chow meal was presented to cafeteria DIO (n ¼ 18) and chow-fed groups (n ¼ 22) after four baseline samples. Only the chow-fed
group showed significant increases in dopamine after the meal. The cafeteria meal that was presented to a subset (n ¼ 8) of the cafeteria DIO group
2.5 h after the regular chow meal resulted in a significant increase in dopamine release (**P < 0.05 between groups, #P < 0.05 within the laboratory
chow-fed group only, and ##P < 0.05 within the cafeteria DIO group only relative to baseline prior to the laboratory chow or cafeteria meal). From
Geiger, B. M., Haburcak, M., Avena, N. M., Moyer, M. C., Hoebel, B. G., & Pothos, E. N. (2009). Deficits of mesolimbic dopamine neurotransmission in rat dietary
obesity. Neuroscience, 159(4), 1193–1199.

agonists and reuptake inhibitors with much more mod- that macronutrient composition can directly affect central
erate effects on body weight continue to be introduced in synaptic plasticity. Recently, a total western diet has been
the market throughout the last decades (Padwal, 2009), introduced that has not only the high fat content of a typ-
although their long-term efficacy and adverse effects ical western diet but also has a modified micronutrient
remain a concern. Overall, central dopamine deficiency content to better match the dietary pattern of the western
(or inefficiency) in obesity seems to be a consistent foun- diet. Initial studies indicate that animals fed this diet are
dation of our understanding of the neurobiology of less susceptible to the excessive weight gain typically
obesity in both animal models and humans, and it seen with high-fat diets. This model provides an opportu-
remains to be established how this deficit can be trans- nity for further investigation into the role of micronutri-
lated to any deficiencies in the perception of food ents in the alteration of CNS pathways involved in
reward. More importantly, it is potentially significant feeding behavior and their effect on the development of
to demonstrate novel ways through which dopamine obesity (Monsanto et al., 2016). As a result of these diet
neurotransmission in the brain can be corrected in order studies, health-care providers may make better dietary
to reduce hyperphagia without potential for addiction and lifestyle recommendations for effective weight loss.
and other serious adverse effects of weight loss-inducing Remarkably, weight management interventions in one
dopaminergic and other drugs. member of a married couple can have crossover benefits
A number of DIO studies have shown further links for the other partner even if the latter is not treated (Gorin
between neurochemistry and diet, including showing et al., 2018).
IV EXAMPLES OF ANIMALS MODELS USED FOR TRANSLATIONAL OBESITY RESEARCH 7
2 Exercise Models in Obesity Research
Along with diet recommendations, exercise is the
most commonly recommended treatment for obesity
by physicians, as increased exercise will tip the energy
balance scales in favor of energy expenditure and weight
loss. To study the effects of exercise, rodents are given
access to a treadmill, and the amount of time they spend
on the treadmill is tracked. In mouse models of diet-
induced obesity, exercise has been linked to not only
lower body weight but also improvements in other
markers of the metabolic syndrome like hyperglycemia
and inflammation (Evans et al., 2014). Exercise also
influences neurotransmitter release and metabolism
that are associated with both energy homeostasis and
hedonic feeding behavior, including that of orexin, lep-
tin, norepinephrine, dopamine, and serotonin. These
effects are seen in both adult animals and animals
immediately post weaning (Greenwood et al., 2011;
Meeusen & De Meirleir, 1995; Novak, Kotz, & Levine,
2006; Obici et al., 2015; Patterson, Bouret, Dunn-
Meynell, & Levin, 2009; Patterson, Dunn-Meynell, &
Levin, 2008; Patterson & Levin, 2008; Thanos et al.,
2010). However, taken to the extreme, excessive exercise
can cause addiction-related changes to the opiate path-
ways in the brain and cause rats to exhibit withdrawal
like symptoms (Kanarek, D’Anci, Jurdak, & Mathes,
2009). Taken together, these findings provide more sup-
port of the effects of exercise on central synaptic plastic-
ity and the use of exercise, particularly in children, as a
safe alternative to pharmacotherapies to combat obe-
sity. In fact, a recent metaanalysis of exercise studies
in obese and overweight children indicates that aerobic
exercise or a combination of aerobic and strength exer-
cises can decrease body weight and adiposity in this
population (Kelley, Kelley, & Pate, 2017).

3 Gut Microbiota Models in Obesity Research


Recent studies have revealed the gut microbiome as
a major contributor to the obesity phenotype in both
mice and humans (Boursi et al., 2018; Hildebrandt
et al., 2009). However, whether alterations in the micro-
biome cause obesity or are the result of obesity is still not
clear (Moran-Ramos, Lopez-Contreras, & Canizales-
FIG. 1.2 Reduced dopamine quantal size in VTA-derived neurons
from P0-P1 obesity-prone pups. (A) Representative amperometric
Quinteros, 2017; Sanmiguel, Gupta, & Mayer, 2015). In
traces from VTA cultures of obesity-resistant (OR) (top) and obesity- general, obesity is linked to lower diversity of microbes
prone (OP) (bottom) neonates. Individual events are shown at higher in the gut, particularly in animal models. There are
resolution. (B) Quantal size distribution in cultures from neonatal OP also differences in the by-products produced by these
and OR animals. Note that the OP distribution is skewed to the left microbes between lean and obese humans and animals.
due to the lack of events in the higher quantal size bins. (C) Quantal size
(left panel) and amplitude (right panel) of stimulated dopamine release
However, the exact significance of these changes is
from VTA-derived neuronal cultures from OP pups (gray bars, n ¼ 110 not completely understood (Moran-Ramos, Lopez-
events) is lower than that in cultures derived from OR pups (black bars, Contreras, et al., 2017). Others have also shown that
n ¼ 182 events). *P < 0.01. From Geiger, B. M., Behr, G. G., Frank, L. E., following laparoscopic sleeve gastrectomy, the gut
Caldera-Siu, A. D., Beinfeld, M. C., Kokkotou, E. G., et al. (2008). Evidence microbiome is altered in patients and these alterations
for defective mesolimbic dopamine exocytosis in obesity-prone rats. FASEB
Journal, 22(8), 2740–2746.
are associated with weight loss, reduced appetite,
and decreased hedonic eating (Sanmiguel et al., 2017).
8 1. TRANSLATING ANIMAL MODELS OF OBESITY AND DIABETES TO THE CLINIC

The molecular mechanisms that contribute to these their groups. After several generations of offering the
changes are not well understood. In animal models, high-fat diet to the animals and selective breeding, the
changes in the microbiota are induced in a variety of offspring of the high weight gainers would spontane-
ways. Diet has significant effects on the composition of ously weigh about 20% more than the offspring of the
the gut microbiota, and studies like the one recently pub- low–weight gaining rats, even when all of the rats were
lished by Moran-Ramos, He, et al. (2017) indicate that fed a normal chow diet (Levin et al., 1997). The rats that
even small changes in the diet of an animal can have sig- gained the excess weight are referred to as diet-induced
nificant effects on the gut microbiota that lead to obese (DIO) or obesity-prone, while the rats that
improvements in the obesity status of the animal. remained leaner are referred to as diet-resistant (DR)
Another approach that has shown promise as a thera- or obesity-resistant animals. In this chapter, we use the
peutic as well is the fecal transfer model. In animals, fecal terms obesity-prone (OP) and obesity-resistant (OR)
matter from lean animals is transferred to obese animals, for selectively bred strains to avoid confusion with our
and changes in the gut microbiota and the overall discussion of models that are developed by exposing
phenotype of the animal have been shown (Moran- the animals to specific diets as part of the study, which
Ramos, Lopez-Contreras, et al., 2017). Based on these were discussed earlier in this chapter. In this section
results, similar studies are being done in humans that the Zucker rat and OP/OR models of obesity as well
so far have shown similar improvements in obesity as others like them are discussed in more detail.
(Marotz & Zarrinpar, 2016; Moran-Ramos, Lopez-
Contreras, et al., 2017; Vrieze et al., 2012). More work 1 Obesity-Prone and Obesity-Resistant Rats
is still necessary in both the preclinical and clinical areas in Translational Research
to identify the molecular mechanisms that are altered in The selectively bred OP/OR strains constitute a model
these studies, particularly as they relate to the gut-brain with many pathophysiological similarities to the human
axis and changes seen in feeding behavior (Kaczmarek, obese state, which is typically viewed as a polygenic
Musaad, & Holscher, 2017). This work can potentially disease. A weight distribution exists between the OP ani-
lead to identification of novel therapeutic targets for mals and the OR animals, similarly to what is seen in
obesity. humans. In the presence of a high-fat diet, these weight
differences are exacerbated with the OP animals gain-
ing much more weight than the OR animals (Levin
et al., 1997). Furthermore, numerous studies have
B Spontaneous and Selectively Bred Animal
shown that a number of different neuropeptides and
Models other CNS proteins and neurotransmitters are altered
In obesity research, some of the most useful animal in these animals, which also corresponds well to the
models have been the spontaneous and selectively bred human condition where multiple neural systems, both
models of obesity and diabetes. These types of models homeostatic and hedonic, have been shown to be
exhibit the phenotype of the disease without specific altered in obese individuals (Berthoud & Morrison,
manipulation of a single gene, in contrast to the genetic 2008; Levin et al., 1997; Levin & Dunn-Meynell, 2002;
models discussed in the next section. For obesity and Levin & Keesey, 1998; Novak et al., 2006). An analysis
many other complex diseases of the CNS, these spontane- of the food reward system in these animals showed
ous models are particularly useful, as the disease will an overall depression of dopamine signaling in both
develop over time as the result of a complex interaction males and females (Fig. 1.3). Most importantly, the par-
of multiple genetic factors. The animal models of obesity ent phenotype affected its offspring similarly to human
discussed in this section are summarized in Table 1.1. obesity. Specifically, attenuation of dopamine release
A spontaneous disease model exhibits the phenotype was evident in neurons that were cultured from the
of the disease without any specific intervention by the dopamine neurons in newborn pups, indicating that
breeder. The Zucker rat is a spontaneous model of obe- the reduction in dopamine was present from birth
sity and hypertension that was first discovered in the and not induced by differences in the animals’ diets
1960s. In contrast, selectively bred models typically arise once they were born (Fig. 1.2; Geiger et al., 2008).
from exposure to a specific environmental influence, and
then the animals are grouped based on their response to 2 Zucker Rats as a Spontaneous Model of Obesity
the influence. The obesity-prone and obesity-resistant The Zucker fatty rat is a widely used model of obesity
rat model is a good example of a selectively bred model that was discovered in the 1960s when a spontaneous
of obesity (Levin et al., 1997). In this model, a colony of mutation in the recessive fa/fa genotype resulted in obe-
Sprague-Dawley rats ate a high-fat diet. The rats that sity and hypertension in these animals (Bray, 1977;
gained the highest and lowest amounts of weight were Zucker & Zucker, 1967). The fa genotype is a missense
separated into two groups and selectively bred within mutation on the leptin receptor gene that renders the gene
Another random document with
no related content on Scribd:
The Project Gutenberg eBook of Chants for the
Boer
This ebook is for the use of anyone anywhere in the United
States and most other parts of the world at no cost and with
almost no restrictions whatsoever. You may copy it, give it away
or re-use it under the terms of the Project Gutenberg License
included with this ebook or online at www.gutenberg.org. If you
are not located in the United States, you will have to check the
laws of the country where you are located before using this
eBook.

Title: Chants for the Boer

Author: Joaquin Miller

Release date: October 16, 2023 [eBook #71889]

Language: English

Original publication: United States: The Whitaker & Ray


Company, 1900

Credits: Tim Lindell, David E. Brown, and the Online Distributed


Proofreading Team at https://www.pgdp.net (This file
was produced from images generously made available
by The Internet Archive/American Libraries.)

*** START OF THE PROJECT GUTENBERG EBOOK CHANTS


FOR THE BOER ***
CHANTS
FOR THE
BOER
By
JOAQUIN MILLER

“And whether on the scaffold high,


Or in the battle’s van,
The fittest place for man to die
Is where he dies for man.”

San Francisco
The Whitaker & Ray Company
(Incorporated)
1900
Copyright, 1900
by
The Whitaker & Ray Company
(Incorporated)
CONTENTS.
TO THE BOERS.
TO YE FIGHTING LORDS OF LONDON TOWN.
MOTHER EGYPT.
ANGLO-SAXON ALLIANCE.
INDIA AND THE BOERS.
AT THE CALEND’S CLOSE.
AS IT IS WRITTEN.
TO OOM PAUL KRUGER.
USLAND TO THE BOERS.
THAT USSIAN OF USLAND.
FIGHT A BOY OF YOUR SIZE.
For the right that needs assistance,
For the wrong that needs resistance,
For the glory in the distance,
For the good that we can do.
Find here not one ill word for brave old England; my first, best friends
were English. But for her policy, her politicians, her speculators, what
man with a heart in him can but hate and abhor them? England’s
best friends to-day are those who deplore this assault on the farmer
Boers, so like ourselves a century back. Could any man be found
strong enough to stay her hand with sword or pen in this mad hour?
That man would deserve her lasting gratitude. This feeling of
abhorrence holds in England as well as here. Take for example the
following from her ablest thinker to a friend in Philadelphia:
“I rejoice that you and others are bent on showing that
there are some among us who think the national honor is
not being enhanced by putting down the weak. Would that
age and ill health did not prevent me from aiding.
“No one can deny that at the time of the Jameson Raid the
aim of the Outlanders and the raiders was to usurp the
Transvaal Government, and he must be willfully blind who
does not see what the Outlanders failed to do by bullets
they hope presently to do by votes, and only those who,
while jealous of their own independence, regard but little
the independence of people who stand in their way, can
fail to sympathize with the Boers in their resistance to
political extinction.
“It is sad to see our Government backing those whose
avowed policy is expansion, which, less politely
expressed, means aggression, for which there is a still
less polite word readily guessed. On behalf of these, the
big British Empire, weapon in hand, growls out to the little
Boer Republic, ‘Do as I bid you.’
“I have always thought that nobleness is shown in treating
tenderly those who are relatively feeble and even
sacrificing on their behalf something to which there is a
just claim. But, if current opinion is right, I must have been
wrong.”
Herbert Spencer.
CHANTS
FOR THE BOER
BY
JOAQUIN MILLER

TO THE BOERS.
“For Freedom’s battles once begun,
Bequeathed from bleeding sire to son,
Though baffled oft, are ever won.”
—BYRON.

The Sword of Gideon, Sword of God


Be with ye, Boers. Brave men of peace
Ye hewed the path, ye brake the sod,
Ye fed white flocks of fat increase
Where Saxon foot had never trod;
Where Saxon foot unto this day
Had measured not, had never known
Had ye not bravely led the way
And made such happy homes your own.

I think God’s house must be such home.


The priestess Mother, choristers
Who spin and weave nor care to roam
Beyond this white God’s house of hers,
But spinning sing and spin again.
I think such silent shepherd men
Most like that few the prophet sings—
Most like that few stout Abram drew
Triumphant o’er the slaughtered Kings.
Defend God’s house! Let fall the crook.
Draw forth the plowshare from the sod
And trust, as in the Holy Book,
The Sword of Gideon and of God;
God and the right! Enough to fight
A million regiments of wrong.
Defend! Nor count what comes of it.
God’s battle bides not with the strong;
And pride must fall. Lo, it is writ!

Great England’s Gold! how stanch she fares


Fame’s wine cup pressing her proud lips—
Her checkerboard of battle squares
Rimmed round by steel-built battleships!
And yet meanwhiles ten thousand miles
She seeks ye out. Well, welcome her!
Give her such welcome with such will
As Boston gave in battle’s whir
That red, dread day at Bunker Hill.

San Francisco, September, 1899.


TO YE FIGHTING LORDS OF LONDON
TOWN.
CHRISTMAS MORNING, 1899.
“The equipment of the Maine hospital ship by our
American cousins warrants us in saying at least that they
wish us well.”

We wish you well in all that’s well,


Would bind your wounds, would clothe, would feed—
Lay flowers where your brave men fell
In desert lands, exalt each deed
Of sacrifice; would beg to lay
White lilies by the gray hearthstone
Where, bowed in black this Christmas day,
She wails her brave dead far away
And weeps, so more than all alone:
Weeps while the chime, the chilly chime,
Drops on her heart, drops all the time
As one might drop a stone.

But you, ye lords and gentlemen


High throned, safe housed at home, fat fed,
When ye say we approve ye, when
Ye say this blood so bravely shed
Is shed with our consent, take care,
Lest Truth may take ye unaware;
Lest Truth be heard despite these chimes.
This hearthstone, brother’s blood that cries
To God is Freedom’s blood. Take care
Lest all sweet earth these piteous times
Not only hate ye for your crimes,
But scorn ye for your lies!

We would forgive could we forget:


We could forget all wrongs we knew
Had ye stayed hand some little yet—
Left to their own that farmer few
So like ourselves that fateful hour
Ye forced our farmers from the plow
To grapple with your tenfold power.
They guessed your greed, we know it now;
And now we ward ye from this hour!
Now, well awake no more we sleep,
But keep and keep and ever keep
To Freedom’s high watchtower.

Not all because our Washington


In battle’s carnage, years and years,
And this same Boer braved ye as one—
Blent blood with blood and tears with tears:
Not all because of kindred blood,
Not all because they built a town
And left such names of true renown.[A]
Not all because of Luther, Huss:
But most because of Brotherhood
In Freedom’s Hall; the holy right
To fight for Home, as freemen fight—
Who Freedom stabs, stabs Us!

This Nation’s heart, say what men may


Who butcher Peace and barter Truth,
Beats true as on its natal day,
Beats true as in its battle-youth,
Beats true to Freedom, true to Truth,
Whatever Tories dare to say.
Of all who fought with Washington
One Arnold was and only one.
Christ chose but twelve, yet one poor soul
Sold God for silver. Ever thus
Some taint, and even so with Us:
But Freedom thrills the whole.

My Lords, ye lead, through Him who died,


Your dauntless millions. Ye are wise
And learned. Ye are, beside,
As God’s anointed in their eyes,
Ye sit so far above their reach.
Such trust! But are ye truly true
To what He taught, to what ye preach,
To those who trust and look to you?
Then why mocked ye that manly Russ,
That august man, that manliest man
That yet has been since time began?
Ye mocked, as ye mock Us!

My Lords, slow paced and somber clad


Ye all will fare to church to-day
And there sit solemn faced and sad
With eyes to book, as if to pray.
And will ye think of Him who came
And lived so poor and died so lorn—
Came in the name of Peace, the name
Of God, that fair first Christmas morn?
My Lords, ye needs must think to-day—
Your eyes bent to the Holy Book
The while the people look and look—
For dare ye try to pray?

And while ye think of Christ the child


Think of the childless mother, she
Whose dead boy has his desert wild,
While yours his Christmas tree;
Think of the mother, far away,
Who sits and weeps with hollow eyes,
Her hungry child that cries and cries
Forlorn and fatherless to-day:
Think of the thousand homes that weep
All desolate, who but for ye
To-day had decked their Christmas tree;
Then fare ye home and—sleep?

[A] Note.—“I thank God there is not a drop of


Saxon blood in my veins. I am a Dutchman; Boer,
if you please.”—Rough-rider Roosevelt, Governor
of New York and heir apparent to the Presidency
of Us.
MOTHER EGYPT.
Dedicated to England on her invasion of North Africa.

Dark browed, she broods with weary lids


Beside her Sphinx and Pyramids,
With low and never-lifted head.
If she be dead, respect the dead;
If she be weeping, let her weep;
If she be sleeping, let her sleep;
For lo, this woman named the stars!
She suckled at her tawny dugs
Your Moses while you reeked in wars
And prowled your woods, nude, painted thugs.

Then back, brave England; back in peace


To Christian isles of fat increase!
Go back! Else bid your high priests bear
The sword and curse the sweet plowshare;
Take down their cross from proud Saint Paul’s
And coin it into cannon-balls!
You tent not far from Nazareth,
Your camps trench where his child-feet strayed.
If Christ had seen this work of death!
If Christ had seen these ships invade!

I think the patient Christ had said,


“Go back, brave men! Take up your dead;
Draw down your great ships to the seas;
Repass the gates of Hercules;
Go back to wife with babe at breast,
And leave lorn Egypt to her rest.”
Or is Christ dead, as Egypt is?
Ah, England, hear me yet again;
There’s something grimly wrong in this—
So like some gray, sad woman slain.

What would you have your mother do?


Hath she not done enough for you?
Go back! And when you learn to read,
Come read this obelisk. Her deed
Like yonder awful forehead is
Disdainful silence. Like to this
What lessons have you writ in stone
To passing nations that shall stand?
Why, years, as hers, will leave you lone
And level as yon yellow sand.

Saint George? Your lions? Whence are they?


From awful, silent Africa.
This Egypt is the lion’s lair;
Beware, brave Albion, beware!
I feel the very Nile should rise
To drive you from this sacrifice.
And if the seven plagues should come?
The red seas swallow sword and steed?
Lo! Christian lands stand mute and dumb
To see thy more than Moslem deed.
ANGLO-SAXON ALLIANCE.
England’s Colonial Secretary, who must bear a great part
of the blame and shame of this Boer war, has said publicly
that there is something like alliance between England and
the United States. Our Secretary of State says there is
nothing of the sort, and we know there is not, nor can be,
until “We, the People,” choose to have it, and that will not
be until this crime against the Boer is forgotten, as well as
Bunker Hill and the Fourth of July.

Alliance! And with whom? For what?


Comes there the skin-clad Vandal down
From Danube’s wilds with vengeance hot?
Comes Turk with torch to sack the town
And wake the world with battle shot?
Come wild beasts loosened from the lair?
No, no! Right fair blue Danube sweeps.
No, no! The Turk, the wild beast sleeps.
No, no! There’s something more than this—
Or Judas’ kiss? Or serpent’s hiss?
There’s mischief in the air!

Alliance! And with whom? For what?


Did we not bear an hundred years
Of England’s hate, hot battle shot,
Blent, ever blent, with scorn and jeers?
And we survived it, did we not?
We bore her hate, let’s try to bear
Her love; but watch her and beware!
Beware the Greek with gifts and fair
Kind promises and courtly praise.
Beware the serpent’s subtle ways—
There’s mischief in the air!

Alliance! And for what? With whom?


She burned our Freedom’s Fane. She spat
Vile venom on the sacred tomb
Of Washington; the while she sat
High throned, fat fed, and safe at home,
And bade slaves hound and burn and slay,
Just as in Africa to-day;
Just as she would, will when she dare
Send sword and torch and once again
Make red the white rim of our main—
There’s mischief in the air!

Alliance! Twice with sword and flame:


Alliance! Thrice with craft and fraud:
And now you come in Freedom’s name.
In Freedom’s name? The name of God!
Go to—the Boers. For shame, for shame!
With wedge of gold you split us twain
Then launched your bloodhounds on the main;
But now, my Lords, so soft, so fair—
How long would this a-lie-ance last?
Just long enough to tie Us fast—
Then music in the air!

You might also like