Discover millions of ebooks, audiobooks, and so much more with a free trial

Only $11.99/month after trial. Cancel anytime.

Translational Regenerative Medicine
Translational Regenerative Medicine
Translational Regenerative Medicine
Ebook2,123 pages17 hours

Translational Regenerative Medicine

Rating: 0 out of 5 stars

()

Read preview

About this ebook

Translational Regenerative Medicine is a reference book that outlines the life cycle for effective implementation of discoveries in the dynamic field of regenerative medicine. By addressing science, technology, development, regulatory, manufacturing, intellectual property, investment, financial, and clinical aspects of the field, this work takes a holistic look at the translation of science and disseminates knowledge for practical use of regenerative medicine tools, therapeutics, and diagnostics. Incorporating contributions from leaders in the fields of translational science across academia, industry, and government, this book establishes a more fluid transition for rapid translation of research to enhance human health and well-being.
  • Provides formulaic coverage of the landscape, process development, manufacturing, challenges, evaluation, and regulatory aspects of the most promising regenerative medicine clinical applications
  • Covers clinical aspects of regenerative medicine related to skin, cartilage, tendons, ligaments, joints, bone, fat, muscle, vascular system, hematopoietic /immune system, peripheral nerve, central nervous system, endocrine system, ophthalmic system, auditory system, oral system, respiratory system, cardiac system, renal system, hepatic system, gastrointestinal system, genitourinary system
  • Identifies effective, proven tools and metrics to identify and pursue clinical and commercial regenerative medicine
LanguageEnglish
Release dateDec 1, 2014
ISBN9780124104570
Translational Regenerative Medicine

Related to Translational Regenerative Medicine

Related ebooks

Medical For You

View More

Related articles

Related categories

Reviews for Translational Regenerative Medicine

Rating: 0 out of 5 stars
0 ratings

0 ratings0 reviews

What did you think?

Tap to rate

Review must be at least 10 words

    Book preview

    Translational Regenerative Medicine - Anthony Atala

    Translational Regenerative Medicine

    Editors

    Anthony Atala, MD

    Julie G. Allickson, PhD

    Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine Winston Salem, USA

    Table of Contents

    Cover image

    Title page

    Copyright

    Dedication

    Contributors

    Chapter 1. The Landscape of Cell Tissues and Organs

    I. The Regenerative Medicine Field

    II. Conclusions

    Disclaimer

    Section I. Cell Banking

    Chapter 2. Landscape of Cell Banking

    I. Introduction: The Field of Cell Banking

    II. Allogeneic and Autologous Cell Banking

    III. Recruitment

    IV. Regulations and Determination of Regulatory Guidance

    V. Registration and Donor Eligibility Screening/Testing

    VI. Current Good Tissue Practice (cGTP)

    VII. Ethics (Personal Data, Health Information, Genetic Analysis, Privately Banked Cells)

    VIII. Financial Aspects

    IX. The Role of Banking in Regenerative Medicine

    X. Conclusion

    Chapter 3. Cell Banking: Process Development and Cell Preservation

    I. Introduction

    II. Product Characterization and Release Specifications

    III. Standard Life Cycle of Banked Cell Therapy Product

    IV. CT Facility

    V. Cleaning and Disinfection

    VI. Environmental Monitoring

    VII. Equipment

    VIII. Critical Reagents and Supplies Management

    Chapter 4. Clinical Development of Placental Mesenchymal Stromal Cells

    Conclusion

    Chapter 5. Translation of Regenerative Medicine Products Into the Clinic in the United States: FDA Perspective

    I. Introduction and Chapter Overview

    II. Brief Legislative History of FDA

    III. Roles of Laws, Regulations, and Guidance

    IV. FDA Organizational Structure and Jurisdictional Processes

    V. Approval Mechanisms and Clinical Studies

    VI. Meetings with Industry, Professional Groups, and Sponsors

    VII. Regulations and Guidance of Special Interest for Regenerative Medicine

    VIII. Preclinical Development Plan

    IX. Clinical Development Plan

    X. Special Topic 1: Current Good Manufacturing Practices

    XI. Special Topic 2: Regulation of Minimally Manipulated, Unrelated Allogeneic Cord Blood

    XII. Special Topic 3: Animal Cell-Based Products for Veterinary Applications

    XIII. Use of Standards in Regenerative Medicine

    XIV. Advisory Committee Meetings

    XV. FDA Regulatory Science Research Initiatives and Critical Path

    XVI. Other Communication Efforts

    XVII. Conclusion

    Chapter 6. Newborn Stem Cell Banking Business Models

    I. Introduction

    II. Cell Biobanking History

    III. Public NSC Biobanking

    IV. Family NSC Biobanking

    V. Operational Execution and Risk Management

    VI. Unit Quality

    VII. Unit Storage Maintenance

    VIII. Risk Mitigation Strategies

    IX. Market Potential

    X. Competition

    XI. Scalability

    XII. Intrinsic Soundness of the Business Model

    XIII. Hybrid NSC Biobanking Business Models

    XIV. The Future of NSC Biobanking

    Acronym

    Section II. Stem Cells and Cell Therapy

    Chapter 7. Cell Therapy Landscape: Autologous and Allogeneic Approaches

    I. Introduction

    II. Comparison of Commercial Potential between Autologous and Allogeneic Cell Therapy

    III. Future Product Commercialization: Will Allogeneic or Autologous Cell Therapy Dominate?

    IV. Conclusion

    List of Abbreviations

    Chapter 8. Stem Cells and Cell Therapy: Autologous Cell Manufacturing

    I. Autologous Therapy

    II. Bone Marrow Aspiration

    III. Manufacturing

    IV. Allogeneic Therapy

    V. Manufacturing

    VI. Summary

    Chapter 9. Overview: Challenges of Process Development for Cellular Therapy

    Chapter 10. Tissue Engineering: Propagation and Potency Evaluation

    I. Introduction: The State of Stem Cell Potency Evaluation

    II. Developing a Reliable Potency Assay with Clinical Relevance

    III. The Future of Stem Cell Potency Enumeration

    Section III. Biomaterials in Regenerative Medicine

    Chapter 11. Biomaterials in Preclinical Approaches for Engineering Skeletal Tissues

    I. Introduction to Skeletal Tissue Engineering (STE)

    II. Biomaterials for Translational Regenerative Medicine

    III. Could Bioreactors Be the Missing Link for Biomechanic Function?

    IV. Scale-Up and Ready to Go Systems

    V. Future Outcomes/Challenges

    List of Acronyms and Abbreviations

    Chapter 12. Biomaterials in Regenerative Medicine: Considerations in Early Process Development

    I. Introduction

    II. Assembling a Design Team

    III. Identifying an Unmet Problem

    IV. Biomaterial-Specific Considerations

    V. Regulatory Challenges

    VI. Conclusion

    VII. Key Points

    List of Acronyms and Abbreviations

    Chapter 13. Biomaterials in Regenerative Medicine: Challenges in Technology Transfer from Science to Process Development

    I. Introduction

    II. Transfer of Biomaterials Technology from Laboratory to Commercial Production: Technical Considerations

    III. Transfer of Technology from Science to Commercial Production: Current Challenges

    IV. Options for Enabling Successful Transition of Technology from Science to Commercialization to Clinical Use

    V. Additional Considerations for Process Development for Biomaterial-Based Products

    VI. Summary

    Further Reading

    Chapter 14. Paracrine Regulation from Tissue Engineered Constructs

    I. Introduction

    II. The Development of Endothelial Cell–Based Paracrine Tissue Engineering Solutions

    III. Designing Paracrine Tissue Engineering Constructs

    IV. Implications of Paracrine Tissue Engineering in Clinical Study Design

    V. Conclusion

    Chapter 15. Creating Commercial Value from Biomaterials

    I. Facing Reality: An Introduction to Translational Medicine and Commercialization

    II. What Features Do You Really Need? Developing the Biomaterial

    III. What Will You Sell? Choosing a Business Model

    IV. Divide and Conquer: A Case Study in Licensing Focused Fields of Use

    V. The Translational Imperative: Deliver Simplicity

    Section IV. Tissue Engineering

    Chapter 16. Manufacturing of Regenerative Medicine Products

    I. Introduction

    II. Manufacturing Process

    III. Manufacturing Facilities and Process Equipment

    IV. Cost of Goods

    V. Good Manufacturing Practices, Good Tissue Practices, and Quality Systems

    VI. Conclusions

    Chapter 17. Regulatory Aspects

    I. Introduction

    II. Regulatory Path

    III. Manufacturing Considerations

    IV. Preclinical Considerations

    V. Clinical Trial Design Considerations

    VI. Developmental Challenges

    VII. Conclusions

    Chapter 18. Global Design for Clinical Trials

    I. Tissue Engineering and Regenerative Medicine

    II. Follow-Up Studies on Clinical Trials of Tissue Engineering

    III. Topics on Scaffold

    IV. Global Design for Clinical Trials of Tissue Engineering

    Section V. Enabling Tools

    Chapter 19. Biomarkers

    I. Introduction

    II. Disease- and Drug-Related Biomarkers

    III. Biomarkers in Drug Development

    IV. Biomarker Requirements

    V. Biomarker Classification and Application

    VI. Discovery of Molecular Biomarkers

    VII. Surrogate End Points and Potential Disadvantages

    VIII. Biomarker in Regenerative Medicine

    XI. Quality Management

    X. Personalized Medicine

    Chapter 20. Translational Animal Models for Regenerative Medicine Research

    I. Introduction

    II. Brief History of Translational Animal Models for Regenerative Medicine Research with an Emphasis on Hematopoietic and Immune System Regeneration

    III. Musculoskeletal Tissue Engineering

    IV. Soft Tissue Regeneration

    V. Ocular and Brain Repair and Regeneration

    VI. Heart Muscle and Vascular Regeneration

    VII. Lung Regeneration

    VIII. Engineered Intestinal, Liver, and Pancreas Regeneration

    IX. Urogenital Repair and Bladder Tissue Engineering

    X. Future Approaches: Stem Cells, Reprogrammed Cells, and Immunodeficient and Humanized Mouse Models for Tissue Regeneration

    XI. Pros and Cons of Translational Animal Models for Regenerative Medicine Research

    Acronyms and Abbreviations

    Chapter 21. Translational Imaging for Regenerative Medicine

    I. Introduction

    II. Contrast Agents

    III. Ultrasound

    IV. X-ray CT

    V. Nuclear Imaging

    VI. Magnetic Resonance Imaging

    VII. Multimodal Imaging

    VIII. Summary

    Section VI. Clinical Aspects of Regenerative Medicine

    Chapter 22. Skin and Skin Appendage Regeneration

    I. Introduction

    II. Epidermis

    III. Hair Follicle and Sebaceous Gland

    IV. Sweat Gland

    V. Future Challenges in Skin Regeneration

    Chapter 23. Clinical Aspects of Regenerative Medicine: Tendon, Ligament, and Joint

    I. Introduction

    II. Platelet-Rich Plasma

    III. Bone Marrow Concentrate

    IV. Discussion

    V. Practical Considerations

    VI. Summary

    Chapter 24. Bone Regeneration

    I. Clinical Importance of Bone Healing

    II. Basic Biology of Bone Healing

    III. Bone Regeneration

    IV. Current and Future Repair Strategies

    V. Conclusions

    Chapter 25. Regenerative Medicine Therapies Using Adipose-Derived Stem Cells

    I. Adipose-Derived Stem Cells for Therapy

    II. Regulatory Process

    III. Current Clinical Trials and Evolving Potential

    IV. Methods of Lipoharvest

    V. Methods of SVF Isolation: Automated versus Manual

    VI. Flow Cytometry Analysis

    VII. Concluding Remarks

    List of Abbreviations

    Chapter 26. Transplantation of Myogenic Cells in Duchenne Muscular Dystrophy Patients: Clinical Findings

    I. Introduction

    II. Cells with Myogenic Capacity: Candidates for Transplantation

    III. Gene Complementation

    IV. Neoformation of Myofibers

    V. Generation of Donor-Derived Satellite Cells

    VI. Control of Acute Rejection

    VII. Potential Future Development in the Control of Acute Rejection

    VIII. The Importance of the Method of Cell Injection

    IX. Potential Future Developments in the Method of Cell Injection

    X. Conclusions

    Chapter 27. Regeneration of the Vascular System

    I. Introduction

    II. Tissue Neovascularization

    III. Bioengineered Blood Vessels

    IV. Summary

    Chapter 28. Hematopoiesis in Regenerative Medicine

    I. Introduction/Historical Perspective

    II. HSC Transplantation for Hematologic Diseases/Disorders

    III. Alternate Sources of HSC

    IV. HSC Transplantation to Induce Immunological Tolerance and Treat Autoimmunity

    V. HSC Transplantation for Diseases of Nonhematopoietic Organs/Tissues

    VI. Summary

    Chapter 29. The Application and Future of Neural Stem Cells in Regenerative Medicine

    I. Establishment of Neural Stem Cells and Induction of Pluripotent Cells for Transplantation

    II. Future Roles of Stem Cell Research

    List of Abbreviations and Acronyms

    Chapter 30. Central Nervous System

    I. Introduction

    II. Regenerative Strategies in the Injured CNS

    III. The Current Landscape of Clinical Trials in the CNS

    IV. Strategies to Address Clinical Challenges for Regenerative Medicine in the CNS

    V. Conclusions and Outlook

    Chapter 31. In situ Tissue Engineering Bone Regeneration in Jaw Reconstruction

    I. The Biology of In situ Tissue Engineering

    II. How the Three Components of the Tissue Engineering Triangle Work to Regenerate Bone

    III. The Clinical Technique Required

    IV. The Prepared In situ Tissue Engineered Bone Graft

    V. Surgery to Place an In situ Tissue Engineered Graft

    VI. Surgery for Mandibular Reconstruction

    VII. Surgery for Maxillary Reconstruction

    VIII. Biologic Activity within an ISTE

    IX. Outcome Analysis: A Three-Cohort Study

    X. Conclusion

    List of Acronyms and Abbreviations

    Chapter 32. Regenerative Medicine for Diseases of the Respiratory System

    I. Introduction

    II. Regenerative Medicine

    III. Tissue Engineering

    IV. Upper Airways: Nasopharynx Vocal Cords and Larynx

    V. Trachea

    VI. Lungs

    VII. Cell Therapy

    VIII. Clinical Translation and Its Barriers

    IX. Mesenchymal Stromal Cells

    X. Mononuclear Cells

    XI. Endothelial Progenitor Cells

    XII. ESCS and iPSCs

    XIII. Other Cell Types

    XIV. Pharmacological Intervention

    XV. Conclusion

    Chapter 33. Renal System

    I. Introduction

    II. Structure and Function of the Kidney

    III. Acute and Chronic Kidney Disease

    IV. Evidence of Normal Kidney Repair in Humans

    V. Regenerative Strategies for Kidney Repair

    VI. Cell Therapy for Renal Failure

    VII. Tissue Engineering Approaches for Renal Failure

    VIII. Conclusion

    Chapter 34. Translational Regenerative Medicine–Hepatic Systems

    I. Introduction: Hepatic Systems

    II. Liver Diseases

    III. Therapies for Liver Diseases

    IV. Liver Transplantation

    V. Cellular Therapies

    VI. Gene Therapy Treatments for Liver Disease

    VII. Liver Bioengineering

    VIII. Liver Assist Devices

    IX. Future Directions

    X. Concluding Remarks

    Chapter 35. Advances in Neo-Innervation of the Gut

    I. Enteric Nervous System: Development and Functions

    II. Enteric Nervous System Disorders: Descriptions and Clinical Treatments

    III. Enteric Neural Cell Transplantation for Treatment of Aganglionic Disorders

    IV. Utilizing Tissue Engineering to Improve Enteric Neural Cell Therapy

    V. Conclusion

    Chapter 36. Genitourinary System

    I. Introduction

    II. Genitourinary System Regeneration

    III. Conclusions and Future Outlook

    Chapter 37. Clinical Aspects of Regenerative Medicine: Immune System

    I. Regenerating the Immune System by Entire or Partial Replacement

    II. Engineering the Immune System with Specialized Component Therapies

    III. Mesenchymal Stem Cells: Specialized Component Therapy Affecting Both Immune Responses and Regeneration

    IV. Conclusions

    Section VII. Translational Aspects of Regenerative Medicine

    Chapter 38. Development of Appropriate Imaging Methods to Trace Cell Fate, Engraftment, and Cell Survival

    I. Introduction

    II. Imaging Approaches

    III. Magnetic Resonance Imaging

    IV. Positron Emission Tomography Imaging

    V. Optical Imaging

    VI. Emerging Approaches

    VII. Selecting a Modality

    VIII. Validation

    IX. Conclusions

    Chapter 39. Gap Analysis to Target Therapies

    I. Introduction

    II. The Pathway

    III. Preclinical Data Packages

    IV. Preclinical Expectations

    V. Product Manufacturing

    VI. Clinical Protocol

    VII. Special Concerns for Cell and Gene Therapy Trials

    Chapter 40. Funding for the Translation of Regenerative Medicines

    I. Introduction

    II. Investment Justification

    III. Global Funding of Regenerative Medicine

    IV. Private Funding of Regenerative Medicine

    V. Stimulating Private Investment

    VI. Creative Government Financing Mechanisms

    VII. Conclusion

    Index

    Copyright

    Academic Press is an imprint of Elsevier

    32 Jamestown Road, London NW1 7BY, UK

    525 B Street, Suite 1800, San Diego, CA 92101-4495, USA

    225 Wyman Street, Waltham, MA 02451, USA

    The Boulevard, Langford Lane, Kidlington, Oxford OX5 1GB, UK

    Copyright © 2015 Elsevier Inc. All rights reserved. Except Chapter 1 which is in the public domain.

    No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical, including photocopying, recording, or any information storage and retrieval system, without permission in writing from the publisher. Details on how to seek permission, further information about the Publisher’s permissions policies and our arrangements with organizations such as the Copyright Clearance Center and the Copyright Licensing Agency, can be found at our website: www.elsevier.com/permissions.

    This book and the individual contributions contained in it are protected under copyright by the Publisher (other than as may be noted herein).

    Notices

    Knowledge and best practice in this field are constantly changing. As new research and experience broaden our understanding, changes in research methods, professional practices, or medical treatment may become necessary.

    Practitioners and researchers must always rely on their own experience and knowledge in evaluating and using any information, methods, compounds, or experiments described herein. In using such information or methods they should be mindful of their own safety and the safety of others, including parties for whom they have a professional responsibility.

    To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume any liability for any injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or operation of any methods, products, instructions, or ideas contained in the material herein.

    ISBN: 978-0-12-410396-2

    British Library Cataloguing in Publication Data

    A catalogue record for this book is available from the British Library

    Library of Congress Catalog Number

    A catalog record for this book is available from the Library of Congress

    For information on all Academic Press publications visit our website at http://store.elsevier.com/

    Typeset by TNQ Books and Journals www.tnq.co.in

    Printed and bound in USA

    Dedication

    This textbook is dedicated to Katherine, Christopher, and Zachary.

    Anthony Atala

    This textbook is dedicated to Brandon and Jason.

    Julie Allickson

    Contributors

    Mehran Abolbashari,     Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA

    Jaimo Ahn,     Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA

    Salem Akel

    St. Louis Cord Blood Bank & Cellular Therapy Laboratory, SSM Cardinal Glennon Children’s Medical Center, St. Louis, MO, USA

    Department of Pediatric, Saint Louis University School of Medicine, St. Louis, MO, USA

    Julie G. Allickson,     Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston Salem, NC, USA

    Graça Almeida-Porada,     Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA

    Judith Arcidiacono,     Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, USA

    Anthony Atala,     Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston Salem, NC, USA

    Patrick Au,     Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, USA

    Danielle Aufiero

    The Orthohealing Center and The Orthobiologic Institute (TOBI), Los Angeles, CA, USA

    David Geffen School of Medicine at UCLA, Los Angeles, CA, USA

    Orthohealing Center, Los Angeles, CA, USA

    Western University of Health Sciences, Pomona, CA, USA

    Touro University, Vallejo, CA, USA

    Pedro M. Baptista

    Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA

    University of Zaragoza, Zaragoza, Spain

    IIS Aragón, CIBERehd, Zaragoza, Spain

    Aragon Health Sciences Institute (IACS), Zaragoza, Spain

    Ronnda L. Bartel,     Aastrom Biosciences, Ann Arbor, MI

    Amelia Bartholomew,     University of Illinois, Department of Surgery, Chicago, IL, USA

    Elona Baum

    Coherus BioSciences, Inc. Redwood City, California

    formerly California Institute for Regenerative Medicine, San Francisco, California, USA

    Angie Botto-van Bemden

    Musculoskeletal Research International (MRI), Ft. Lauderdale, FL, USA

    Clinical Research Experts (CRE), Ft. Lauderdale, FL, USA

    Florida International University, Miami, FL, USA

    Khalil N. Bitar

    Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA

    Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Winston-Salem, NC, USA

    Lynne Boxer,     Center for Veterinary Medicine, FDA, Rockville, MD, USA

    Matthew P. Brown,     Founder Shama Consulting

    Heather L. Brown,     Vice President Scientific Medical Affairs, Cord Blood Registry, San Bruno, CA, USA

    Stephanie J. Bryant,     Department of Chemical and Biological Engineering, College of Engineering and Applied Science, University of Colorado, Boulder, CO, USA

    Pedro P. Carvalho

    3B’s Research Group, Department of Polymer Engineering, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal

    ICVS/3B’s – PT Government Associate Laboratory, Braga/Guimarães, Portugal

    Prafulla Chandra,     Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA

    John R. Chapman,     President of Stem Cell Partners & Adjunct Professor of Biology, California State University Sacramento, USA

    Shreyasi Das,     Sanford-Burnham Medical Research Institute, San Diego, CA, USA

    Daniel B. Deegan,     Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA

    Abritee Dhal,     Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA

    Albert D. Donnenberg

    University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA

    Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA

    Matthew B. Durdy,     Cell Therapy Catapult, Guy’s Hospital, Great Maze Pond, London, UK

    Charles N. Durfor,     Center for Devices and Radiological Health, FDA, Silver Spring, MD, USA

    Elazer R. Edelman

    Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA

    Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA

    Donald Fink,     Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, USA

    Steven Fischkoff,     Celgene Cellular Therapeutics, Warren, NJ, USA

    Joyce L. Frey-Vasconcells,     Frey-Vasconcells Consulting, LLC, Sykesville, MD, USA

    Tobias Führmann,     Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada

    Carmen Gacchina Johnson

    Commissioner’s Fellowship Program, Office of the Commissioner, U.S. Food and Drug Administration, Silver Spring, MD, USA

    Center for Devices and Radiological Health, FDA, Silver Spring, MD, USA

    Or Gadish,     Harvard-MIT Program in Health Sciences and Technology, & Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA

    Sanjiv S. Gambhir

    Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, CA, USA

    Bioengineering, Materials Science & Engineering, Bio-X, Stanford University, Stanford, CA, USA

    Adrian P. Gee,     Center for Cell & Gene Therapy, Baylor College of Medicine, Houston, Texas, USA

    Manuela E. Gomes

    3B’s Research Group, Department of Polymer Engineering, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal

    ICVS/3B’s – PT Government Associate Laboratory, Braga/Guimarães, Portugal

    Kurt D. Hankenson,     Department of Small Animal Clinical Sciences College of Veterinary Medicine; and Department of Physiology, Colleges of Natural Sciences and Osteopathic Medicine

    Robert J. Hariri,     Celgene Cellular Therapeutics, Warren, NJ, USA

    Heather C. Hatcher,     Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston Salem, NC, USA

    Mohammad Heidaran,     Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, USA

    Ralf Huss

    University of Munich, Munich, Germany

    Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA

    Apceth GmbH & Co. KG, Munich, Germany

    Definiens AG, Munich, Germany

    John Hyde,     Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, USA

    Yoshito Ikada,     Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan

    Deepak Jain,     Bioprocess Research & Development, Manufacturing and Technical Operations, Tengion, Inc., Winston-Salem, NC, USA

    Paul A. Jain,     University of California-San Diego Medical Center, San Diego Veterans Affair Medical Center, San diego, CA, USA

    Jesse V. Jokerst,     Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, CA, USA

    Philipp Jungebluth,     Department of Clinical Science, Intervention and Technology, Division of Ear, Nose and Throat, Advanced Center for Translational Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden

    Eve Kandyba

    Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA

    Department of Pathology, University of Southern California, Los Angeles, CA, USA

    David S. Kaplan,     Center for Devices and Radiological Health, FDA, Silver Spring, MD, USA

    Safa Karandish,     Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, USA

    F. Kurtis Kasper,     Department of Bioengineering, Rice University, Houston, TX, USA

    Sneha S. Kelkar

    Wake Forest–Virginia Tech School of Biomedical Engineering and Sciences, Wake Forest University Health Sciences, Winston-Salem, NC, USA

    Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA

    Norma Kenyon,     Diabetes Research Institute, Miami, FL, USA

    Krzysztof Kobielak

    Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA

    Department of Pathology, University of Southern California, Los Angeles, CA, USA

    Jesse Kramer,     Vice President Scientific Medical Affairs, Cord Blood Registry, San Bruno, CA, USA

    Sang Jin Lee,     Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA

    Mark H. Lee,     Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, USA

    Yvonne Leung

    Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA

    Department of Pathology, University of Southern California, Los Angeles, CA, USA

    Mei Ling Lim,     Department of Clinical Science, Intervention and Technology, Division of Ear, Nose and Throat, Advanced Center for Translational Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden

    Neil J. Littman,     California Institute for Regenerative Medicine, San Francisco, California, USA

    Paolo Macchiarini,     Department of Clinical Science, Intervention and Technology, Division of Ear, Nose and Throat, Advanced Center for Translational Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden

    Nafees N. Malik

    Cell Therapy Catapult, Guy’s Hospital, Great Maze Pond, London, UK

    Asklepian Consulting, Birmingham, UK

    Institute of Biotechnology, Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK

    Brenda K. Mann,     SentrX Animal Care, Salt Lake City, UT, USA

    Kacey G. Marra

    Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA

    McGowan Institute of Regenerative Medicine, Pittsburgh, PA, USA

    Robert E. Marx,     Chief Division of Oral and Maxillofacial Surgery, University of Miami Miller School of Medicine, Miami, FL, USA

    Lina Mastrangelo,     Sanford-Burnham Medical Research Institute, San Diego, CA, USA

    Brent McCright,     Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, USA

    Richard McFarland,     Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, USA

    Michael Mendicino

    Commissioner’s Fellowship Program, Office of the Commissioner, U.S. Food and Drug Administration, Silver Spring, MD, USA

    Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, USA

    Antonios G. Mikos,     Department of Bioengineering, Rice University, Houston, TX, USA

    Nikolaos Mitrousis,     Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada

    Aaron M. Mohs

    Wake Forest–Virginia Tech School of Biomedical Engineering and Sciences, Wake Forest University Health Sciences, Winston-Salem, NC, USA

    Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA

    Department of Cancer Biology, Wake Forest University Health Sciences, Winston-Salem, NC, USA

    Thomas Moore,     Founder Shama Consulting

    Emma C. Moran,     Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA

    Walter Niles,     Sanford-Burnham Medical Research Institute, San Diego, CA, USA

    Guoguang Niu,     Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, Winston–Salem, NC, USA

    Masashi Nomi,     Department of Urology, Suma-Ku, Kobe Children’s Hospital, Kobe, Japan

    Tamara Nunez,     University of Illinois, Department of Surgery, Chicago, IL, USA

    Robert Perry,     Athersys, Inc., Cleveland, Ohio, USA

    Robert P. Pfotenhauer,     Vice President Scientific Medical Affairs, Cord Blood Registry, San Bruno, CA, USA

    Christopher D. Porada,     Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA

    Kavitha Premenand,     University of Illinois, Department of Surgery, Chicago, IL, USA

    Glenn D. Prestwich,     Department of Medicinal Chemistry, The University of Utah, Salt Lake City, Utah, USA

    Shreya Raghavan

    Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA

    Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Winston-Salem, NC, USA

    Mahendra Rao,     NIH Center for Regenerative Medicine, Bethesda, MD, USA

    Anthony Ratcliffe,     Synthasome, Inc., San Diego, CA, USA

    Stephen Rego,     Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA

    Rui L. Reis

    3B’s Research Group, Department of Polymer Engineering, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal

    ICVS/3B’s – PT Government Associate Laboratory, Braga/Guimarães, Portugal

    Ivan N. Rich,     HemoGenix, Inc, Colorado Springs, CO, USA

    Márcia T. Rodrigues

    3B’s Research Group, Department of Polymer Engineering, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal

    ICVS/3B’s – PT Government Associate Laboratory, Braga/Guimarães, Portugal

    J. Peter Rubin

    Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA

    McGowan Institute of Regenerative Medicine, Pittsburgh, PA, USA

    Steven Sampson

    The Orthohealing Center and The Orthobiologic Institute (TOBI), Los Angeles, CA, USA

    David Geffen School of Medicine at UCLA, Los Angeles, CA, USA

    Orthohealing Center, Los Angeles, CA, USA

    Western University of Health Sciences, Pomona, CA, USA

    Touro University, Vallejo, CA, USA

    Etai Sapoznik

    Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, Winston–Salem, NC, USA

    Virginia Tech – Wake Forest University School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, VA, USA

    John G. Sharp,     Department of Genetics, Cell Biology & Anatomy, University of Nebraska Medical Center, Omaha, NE, USA

    Molly S. Shoichet

    Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada

    Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada

    Department of Chemistry, University of Toronto, Toronto, ON, Canada

    Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada

    Daniel Skuk,     Neurosciences Division—Human Genetics, CHUQ Research Center—CHUL, Quebec, QC, Canada

    Evan Y. Snyder,     Sanford-Burnham Medical Research Institute, San Diego, CA, USA

    Shay Soker

    Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, Winston–Salem, NC, USA

    Virginia Tech – Wake Forest University School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, VA, USA

    Sita Somara,     Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA

    Tom Spencer,     Process Development and Manufacturing, Tengion, Inc., Winston-Salem, NC, USA

    Suzanne Stewart,     Department of Clinical Studies-New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA, USA

    Premenand Sundivakkam,     University of Illinois, Department of Surgery, Chicago, IL, USA

    Erszebet Szilagyi,     University of Illinois, Department of Surgery, Chicago, IL, USA

    Alexander M. Tatara,     Department of Bioengineering, Rice University, Houston, TX, USA

    Brian Tobe

    Sanford-Burnham Medical Research Institute, San Diego, CA, USA

    Department of Psychiatry, Veterans Administration Medical Center, San Diego, CA, USA

    Jacques P. Tremblay,     Neurosciences Division—Human Genetics, CHUQ Research Center—CHUL, Quebec, QC, Canada

    Alan O. Trounson,     Richie Centre, Monash University, Monash Medical Centre, Clayton, Victoria, Australia

    Anup Tuladhar,     Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada

    Lori Tull,     Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, USA

    Jolene E. Valentin,     Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA

    Dipen Vyas,     Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA

    Zhan Wang,     Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, Winston–Salem, NC, USA

    Alicia Winquist,     Sanford-Burnham Medical Research Institute, San Diego, CA, USA

    Celia Witten,     Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, USA

    Mark E.K. Wong,     Department of Oral and Maxillofacial Surgery, University of Texas Health Science Center at Houston, Houston, TX, USA

    James J. Yoo,     Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA

    Diana Yoon

    Commissioner’s Fellowship Program, Office of the Commissioner, U.S. Food and Drug Administration, Silver Spring, MD, USA

    Center for Devices and Radiological Health, FDA, Silver Spring, MD, USA

    Elie Zakhem,     Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA

    Joao Paulo Zambon,     Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA

    Chapter 1

    The Landscape of Cell Tissues and Organs

    Mahendra Rao     NIH Center for Regenerative Medicine, Bethesda, MD, USA

    Abstract

    The regenerative medicine field is active and growing. It should not be considered as a unified field, but rather as a set of subfields that focus on different cells and different indications and are regulated by diverse regulatory pathways. The field remains united conceptually in that the players are all motivated to use cells, engineered cells, cells in combination with devices, and cell derivatives for treating disorders. As with any new field, several business models are being explored, and innovators are looking at different ways to offer services and to obtain returns on their investments. The regulatory authorities have had to scramble to keep up with these innovative breakthroughs, and academic investigators have developed ever more sophisticated ways to combine cells with biomaterials to generate complex three-dimensional structures to replace failing tissue. The field is dynamic, and new breakthroughs may change the field or accelerate existing trends.

    Keywords

    hematopoietic stem cells; mesenchymal stem cells; neural stem cells; regenerative medicine; Stem cell

    Chapter Outline

    I. The Regenerative Medicine Field 3

    II. Conclusions 7

    Disclaimer 8

    References 8

    I. The Regenerative Medicine Field

    The dream of curing illness and injury by transplanting organs, bone, and other tissue is probably as old as the history of healing, with the first recorded attempts dating back to the Middle Ages [1–3] when tissue flaps were mobilized for wound repair. The term Regenerative Medicine is widely attributed to having first been coined by William Haseltine (founder of Human Genome Sciences), although the term was first found in a 1992 article on hospital administration by Leland Kaiser. Kaiser’s paper closes with a series of short paragraphs on future technologies that will have an impact on hospitals. One such paragraph had Regenerative Medicine as a bold print title and went on to state, A new branch of medicine will develop that attempts to change the course of chronic disease and in many instances will regenerate tired and failing organ systems. The regenerative medicine field as I see it is summarized in Figure 1. The field, as the National Institutes of Health (NIH) sees it, is summarized in a report [4] that states that regenerative medicine is the application of treatments developed to replace tissues damaged by injury or disease. These treatments may involve the use of biochemical techniques to induce tissue regeneration directly at the site of damage or the use of transplantation techniques using differentiated cells or stem cells, either alone or as part of a bioartificial tissue.

    Successful transplantation of bone, skin, and corneas came first, with advances made between 1900 and 1920. The establishment of the U.S. Navy Tissue Bank in 1949 gave the nation its first bone and tissue processing and storage facility, and since then we have seen the birth of the bone marrow registries, cord blood and other tissue banks, and the Embryonic Stem Cells (ESC) and Induced Pluripotent Stem Cells (IPSC) banking initiatives. Table 1 provides an abbreviated timeline of some major milestones in tissue and organ transplants. Perhaps an important milestone is that by the 1990s, over half a million tissues had been transplanted and had benefitted hundreds of thousands of individuals. Equally important, the field has allowed us to standardize processes to collect, process, store, and distribute tissue and to understand the difficulties in solving the immune rejection phenomenon.

    Other breakthroughs in immune suppression, such as cyclosporine treatment, allowed the development of bone marrow transplants [5]; this has led to treatment for a wide variety of disorders, and well over 25,000 bone marrow transplantations occur each year. The utility of bone marrow led to the search for other sources of blood cells, and two different findings have sculpted the field. One was the finding that mobilization simplifies the process of obtaining CD34-positive cells as compared to isolating bone marrow [6], and the other was that finding that cord and placenta contained a large number of engraftable marrow-like blood stem cells [7]. This observation has led to the birth of an entire industry dedicated to the collection and storage of cord blood and its use as an alternative source, supplement, or better substitute for marrow, particularly in children. A recent milestone reached was that the number of cord transplantations in children have exceeded those of bone marrow [8].

    It was Friedenstein and coworkers, in a series of seminal studies in the 1960 and 1970s [9], who showed that the osteogenic potential, of BM cells, was associated with a minor subpopulation of cells in the bone marrow isolate. These cells were distinguishable from the majority of hematopoietic cells by their rapid adherence to tissue culture vessels and by the fibroblast-like appearance of their progeny in culture, pointing to their origin from the stromal compartment of BM. The currently popular albeit perhaps inaccurate term mesenchymal stem cells (MSCs) was first coined in 1991 by Dr Arnold Caplan (Paolo Bianco et al.). Work by Darwin Prockop and others (Phinney, Pittenger) further defined the cells and their multilineage capability. The ability to grow an autologous cell relatively easily and to obtain very large numbers of cells that appeared to be able to perform a wide variety of functions (Figure 2) has led to the birth of an entire subfield of regenerative medicine. More than 1000 trials have been run; more than 50 companies offering some variant of a mesenchymal cell are in existence; and commercial Food and Drug Administration (FDA)–approved products that use MSC are available to treat some diseases.

    Figure 1   The overall field of regenerative medicine is broad and can be divided into several subfields based on whether cells, tissues or combination materials are used.

    Figure 2   The multiple different ways mesenchymal stem cell like cells are used is summarized. Both autologous and allogeneic cells are available and can be used in most indications. The immune modulatory role of MSC in particular is being explored.

    Table 1

    Historical Highlights and Milestones

    This table lists some milestones in using cells and tissue for transplants.

    Modified from http://www.mtf.org/news_history_of_transplantation.html.

    The ability to obtain HSC, cord blood–derived HSC, and MSC has allowed investigators to explore isolatable cells in other fields as well. Corneal Limbal cells found a niche use, and since the 1990s stem cells or progenitor cells or, perhaps more accurately, cells with a finite but impressive self-replicating potential, can be isolated from virtually all tissues. This has led to a further expansion of the regenerative medicine field (Figure 3).

    Irrespective of how one defines the field or attributes the name, what is clear is that the field is growing by leaps and bounds due to the convergence of several different advances and recent breakthroughs (Figure 4). The Nobel Prize–winning work of Dr. Guerdon and Dr. Yamanaka [10,11] and the implementation of new methods of making human PSC [12] engineering pluripotent cells that further build on the Nobel prize-winning work of Dr. Capecchi and others [13,14] have further fueled the growth of this field. The past decade has seen an exponential increase in the products approved, the number of clinical trials initiated (www.cinicaltrials.gov), the number of biotechnology companies incorporated, and the number of papers published [15,16]. These developments have led to the treatment of hundreds of thousands of patients and the creation of entire new subfields fields such as that of induced pluripotence [17].

    Making ESC from human cells and the demonstration that these cells are a unique population of cells that do not undergo senescence has meant that, in theory, every investigator had access to the same cell type and studies could be compared without the confounding influence of allelic variability, laboratory-to-laboratory variations, and other cell culture artifacts. The generation of iPSC in addition to forcing us to reevaluate our understanding of development in essence brought ESC technology to the masses, and for investigators working in the translation field suggested that the immune issue for transplantation could be solved in an elegant way.

    Parallel advances in Next-GEN sequencing, which have exponentially reduced the cost and the number of cells required for analysis while increasing the variety of information that can be processed, have provided a depth of data that has allowed rapid analysis of single genomes and cell states [18,19]. Today it is very possible to sequence the entire genome, and to obtain an epigenome methylation and miRNA profile with as few as 10,000  cells at a cost that is well within the reach of an average laboratory. Such well-characterized pluripotent cell populations can then be readily grown and differentiated into multiple cell types using cost-saving bioreactor technology that has been optimized over the years for bioproduction and antibody generation, and that is now being adapted to primary cell culture. Perhaps equally important and, in my opinion a critical development, was our ability to preserve cells indefinitely by holding them frozen for years. Although we often take this for granted, one realizes how vital this is when it is not possible for us to do so. The entire skin therapy field has been shaped by the fact that cell sheets have to be shipped live and thus manufactured to demand, changing the entire cost structure of that industry. Success in storing hematopoietic stem cells, on the other hand, has shaped that industry differently.

    Figure 3   The different kinds of cells being considered for therapy are listed.

    Figure 4   The field of regenerative medicine is changing because of the many breakthroughs and advances in different scientific disciplines can be synergistically utilized.

    Complementary advances in engineering cells, in particular homologous recombination and its variants using ZFNs [20], TALENS [21] or CAS9 systems [22], now allow us to take well-characterized pluripotent cells that have been comprehensively annotated and make single-gene (or multiple-gene) changes to model complex diseases or to repair cells for therapy. This level of unprecedented control over a system with high efficiency and high fidelity allows us to address problems that were simply intractable in the past.

    Our ability to make cells jump through hoops has been complemented by advances in tissue engineering, biomimetic scaffolds, the development of thermoreversible gels that work at physiological temperatures, and three-dimensional tissue printing that allows us to mold structures using biological grade material, seed them with cells, and begin to develop two- and three-dimensional organized structures that can be used for therapy [23–25].

    Table 2

    Abbreviated List of Companies that Have a Market Cap of Greater than $20M

    Note: Several of these companies are close to billion-dollar companies even though few have an approved product on the market. ALS, Alzheimer’s disease; CLI, critical limb ischemia; CNS, central nervous system; EMA, European Medicines Agency; ESC, embryonic stem cells; GVD, graft-versus-host disease; IBD, inflammatory bowel disease; MSC, mesenchymal stromal/stem cells; NA, Not Applicable.

    The stem cell field has been shaped not just by technology and scientific breakthroughs but also by patents and regulations governing the delivery of cell-based therapy as well as by the enormous expectations that people have for this novel type of therapy [26–28]. Likewise, the field of regenerative medicine has been strongly influenced by regulatory authorities who have imposed different requirements in different countries [29–31]. The regulations have led to different foci of activity in different countries and, by extension, the players who implement these technologies. Overall, the field has shown some success, and there are several cell and tissue therapy products on the market, several companies have been set up, and more than 10 have a market capitalization of great than $20  million, with some being close to billion-dollar companies (Table 2). Several new discoveries have been made that have led to new spin-offs, and hundreds of patients have been treated. New business models for autologous therapy are being developed, and hundreds of clinical trials have been initiated.

    II. Conclusions

    The regenerative medicine field is active and growing. It should not be considered as a unified field but, rather, as a set of subfields that focus on different cells and different indications and that are regulated by diverse regulatory pathways. The field remains united conceptually in that the players are all motivated to use cells, engineered cells, cells combined with devices, and cell derivatives for treating disorders. As with any new field, several business models are being explored, and innovators are looking at different ways to offer services and to obtain returns on their investments. The regulatory authorities have had to scramble to keep up with these innovative breakthroughs, and have succeeded in reining in some of the excesses and hype in the field. Given the large number of companies and the ongoing investments and the recent approval of products in the United States and Korea, it is likely that the field will continue to grow.

    I also remind investigators that the field is in a state of flux, and new discoveries have the possibility of changing the field yet again. As with any prediction, it is far more likely that I will be wrong than right, but nevertheless one can perhaps make some cautious predictions [32]. It seems to this writer that the gene engineering breakthroughs coupled with the ability to make differentiated cells of various kinds will allow functional cures. Perhaps the earliest success will come in the hematopoietic system or in the retina, where viral delivery of a missing gene product has already shown success. Using safe-harbor technologies and strategies to prevent silencing and regulating gene expression in immune-matched cells that are differentiated into a tissue or organ phenotype will allow controlled and regulated therapy. Another breakthrough that I believe will change the field is the ability to make more complex three-dimensional structures. Thus far we have been limited by our inability to construct a true vasculature that will allow nutrient delivery in synthetic constructs that are more than eight or 10 cell layers thick. Keeping organ structures alive while a vasculature develops has been difficult as well. Current work in angiogeneisis, the ability to print organs layer by layer, and the isolation of endothelial cells in large numbers suggest that we are close. A third breakthrough that builds on the work on iPSC is the idea of direct transdifferentiation using readily available sources of adult cells and directing their differentiation into a difficult-to-obtain cell type. Work by several investigators has suggested that this may be possible, and in some cases may be possible in vivo.

    Disclaimer

    The opinions expressed in this article are entirely my own and do not reflect the opinions or policy of the National Institutes of Health.

    References

    [1] http://stemcells.nih.gov/info/scireport/pages/2006report.aspx, published in 2006.

    [2] Dinsmore C.E, ed. A history of regeneration research: milestones in the evolution of a science. Cambridge University Press; 1991.

    [3] Berthiaume F, Maguire T.J, Yarmush M.L. Tissue engineering and regenerative medicine: history, progress, and challenges. Annu Rev Chem Biomol Eng. 2011;2:403–430. doi: 10.1146/annurev-chembioeng-061010-114257.

    [4] Polykandriotis E, Popescu L.M, Horch R.E. Regenerative medicine: then and now–an update of recent history into future possibilities. J Cell Mol Med. October, 2010;14(10):2350–2358. doi: 10.1111/j.1582-4934.2010.01169.x.

    [5] de la Morena M.T, Gatti R.A. A history of bone marrow transplantation. Hematol Oncol Clin North Am. February, 2011;25(1):1–15. doi: 10.1016/j.hoc.2010.11.001.

    [6] Jansen J, Hanks S, Thompson J.M, Dugan M.J, Akard L.P. Transplantation of hematopoietic stem cells from the peripheral blood. J Cell Mol Med. 2005 January–March;9(1):37–50.

    [7] Gluckman E. History of cord blood transplantation. Bone Marrow Transpl. 2009;44:621–626. doi: 10.1038/bmt.2009.280 published online October 5, 2009.

    [8] Rao M, Ahrlund-Richter L, Kaufman D.S. Concise review: cord blood banking, transplantation and induced pluripotent stem cell: success and opportunities. Stem Cells. January, 2012;30(1):55–60. doi: 10.1002/stem.770.

    [9] Friedenstein A.J. Stromal-hematopoietic interrelationships: Maximov’s ideas and modern models. Haematol Blood Transfus. 1989;32:159–167.

    [10] Gurdon J.B, Byrne J.A. The first half-century of nuclear transplantation. Proc Natl Acad Sci. 2003;100(14):8048. doi: 10.1073/pnas.1337135100.

    [11] Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. November 30, 2007;131(5):861–872.

    [12] A review of the methods for human iPSC derivation. Malik N, Rao M.S. Methods Mol Biol. 2013;997:23–33. doi: 10.1007/978-1-62703-348-0_3.

    [13] Thomas K.R, Capecchi M.R. Site-directed mutagenesis by gene targeting in mouse embryo-derived stem cells. Cell. November 6, 1987;51(3):503–512.

    [14] Altering the genome by homologous recombination. Capecchi M.R. Science. June 16, 1989;244(4910):1288–1292 [Review].

    [15] http://www.stemcellresources.org/who_companies.html.

    [16] Sanberg PR, Borlongan CV. The proliferation and differentiation of stem cell journals. Stem Cell Rev Rep. doi:10.1007/s12015-010-9181-y

    [17] Yamanaka S. Induced pluripotent stem cells: past, present, and future. Cell Stem Cell. June 14, 2012;10(6):678–684. doi: 10.1016/j.stem.2012.05.005.

    [18] Metzker M.L. Sequencing technologies - the next generation. Nat Rev Genet. January, 2010;11(1):31–46. doi: 10.1038/nrg2626 Epub December 8, 2009.

    [19] Morozova O, Marra M.A. Applications of next-generation sequencing technologies in functional genomics. Genomics. November, 2008;92(5):255–264.

    [20] Carroll D. Genome engineering with zinc-finger nucleases. Genetics. August, 2011;188(4):773–782. doi: 10.1534/genetics.111.131433.

    [21] Sun N, Zhao H. Transcription activator-like effector nucleases (TALENs): a highly efficient and versatile tool for genome editing. Biotechnol Bioeng. July 2013;110(7):1811–1821. doi: 10.1002/bit.24890 Epub April 7, 2013.

    [22] Gaj T, Gersbach C.A, Barbas 3rdC.F. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering S0167–7799(13)00087-5. Trends Biotechnol. May 8, 2013 doi: 10.1016/j.tibtech.2013.04.004 Epub ahead of print.

    [23] Kehoe S, Zhang X.F, Boyd D. FDA approved guidance conduits and wraps for peripheral nerve injury: a review of materials and efficacy. Injury. May, 2012;43(5):553–572.

    [24] Zorlutuna P, Vrana N.E, Khademhosseini A. The expanding world of tissue engineering: the building blocks and new applications of tissue engineered constructs. IEEE Rev Biomed Eng. 2013(6):47–62. doi: 10.1109/RBME.2012.2233468 Epub December 20, 2012.

    [25] Takagi R, Yamato M, Kanai N, Murakami D, Kondo M, Ishii T, et al. Cell sheet technology for regeneration of esophageal mucosa. World J Gastroenterol. October 7, 2012;18(37):5145–5150. doi: 10.3748/wjg.v18.i37.5145.

    [26] Vrtovec K.T, Scott C.T. Patenting pluripotence: the next battle for stem cell intellectual property. Nat Biotechnol. April, 2008;26(4):393–395. doi: 10.1038/nbt0408-393.

    [27] Sipp D, Turner L. Stem cells. U.S. regulation of stem cells as medical products. Science. December 7, 2012;338(6112):1296–1297. doi: 10.1126/science.1229918.

    [28] Ikegaya H. Bioethics: tighten up Japan’s stem-cell practices. Nature. September 6, 2012;489(7414):33. doi: 10.1038/489033c.

    [29] Caulfield T, McGuire A. Athletes’ use of unproven stem cell therapies: adding to inappropriate media hype? Mol Ther. September, 2012;20(9):1656–1658. doi: 10.1038/mt.2012.172.

    [30] Allison M. Hemacord approval may foreshadow regulatory creep for HSC therapies. Nat Biotechnol. April 10, 2012;30(4):304. doi: 10.1038/nbt0412-304.

    [31] Carpenter M.K, Couture L.A. Regulatory considerations for the development of autologous induced pluripotent stem cell therapies. Regen Med. July 2010;5(4):569–579. doi: 10.2217/rme.10.55.

    [32] Rao M. Stem Cell for Therapy. Tissue engineering and regenerative medicine. 2013;10(5):223–229.

    Section I

    Cell Banking

    Outline

    Chapter 2. Landscape of Cell Banking

    Chapter 3. Cell Banking: Process Development and Cell Preservation

    Chapter 4. Clinical Development of Placental Mesenchymal Stromal Cells

    Chapter 5. Translation of Regenerative Medicine Products Into the Clinic in the United States: FDA Perspective

    Chapter 6. Newborn Stem Cell Banking Business Models

    Chapter 2

    Landscape of Cell Banking

    Heather C. Hatcher, Anthony Atala,  and Julie G. Allickson     Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston Salem, NC, USA

    Abstract

    As the aging population grows in the United States and globally, regenerative medicine has become an important healthcare sustainability goal, as it has the potential to save billions of dollars in healthcare costs. One important aspect of regenerative medicine is cellular therapy, where the cellular component used in the construction of organs and tissue is extremely vital. Banking stem cells has generally been seen in the bone marrow and cord blood transplant fields for hematopoietic diseases; however, within the last decade regenerative medicine has expanded the use of banked stem cells to neurodegenerative, cardiac, and musculoskeletal diseases. The biobanking field for clinical use includes a public-unrelated/universal-donor model as well as an autologous-source model consisting of privately storing one's own cells. Banking cells for public use or to establish cell lines that could be used for multiple patients may be associated with financial savings, as the execution of federal regulations on one donor cell line would facilitate sourcing individual donors for each patient. With private banking or banking one's own cells, the threat of immune rejection is diminished; however, the time and cost required is a challenge compared to using cell lines or a universal donor. Taking into account the regulatory considerations, time constraints of patient treatment, and financial considerations, both public and private cell banking will play significant roles in the future of translational regenerative medicine.

    Keywords

    Clinical application; Public and private cord blood banks; Regenerative medicine; Translational medicine; Umbilical cord blood

    Chapter Outline

    I. Introduction: The Field of Cell Banking 13

    II. Allogeneic and Autologous Cell Banking 13

    III. Recruitment 14

    IV. Regulations and Determination of Regulatory Guidance 14

    V. Registration and Donor Eligibility Screening/Testing 15

    VI. Current Good Tissue Practice (cGTP) 16

    VII. Ethics (Personal Data, Health Information, Genetic Analysis, Privately Banked Cells) 16

    VIII. Financial Aspects 17

    IX. The Role of Banking in Regenerative Medicine 17

    X. Conclusion 18

    References 18

    I. Introduction: The Field of Cell Banking

    In recent years, cell banking activities have expanded worldwide. In 2010, nearly 17,000 products derived from bone marrow, peripheral blood stem cells (BSCs), and cord blood units (CBUs) were available worldwide for allogeneic transplantation to unrelated patients suffering oncologic, genetic, hematologic, and immunodeficiency disorders [1]. Human umbilical cord blood (UCB) has gained increasing importance as a source of BSCs and mesenchymal stem cells (MSCs) [2,3]. Clinical experience over the last two decades has shown that cord blood (CB) is a viable source for BSCs in the field of unrelated hematopoietic blood-stem-cell transplantation [4]. CB has significant advantages as a source of hematopoietic stem cells, including convenience to the donor, reduced risk of graft-versus-host disease, up to two human leukocyte antigen (HLA) mismatches, and almost immediate availability [5]. In 1989, the first CB transplantation was reported in a boy with Fanconi anemia, and the first series of CB transplantations from related and unrelated donors was reported in the mid-1990s (for review [6]). In the early 1990s, the first unrelated CB banking programs were started at the Eurocord/NetCord Bank in Düsseldorf and the New York Blood Center [7,8]. By 2010, the global inventory of CB available for transplantation was approximately 600,000 units, stored in more than 140 CB banks worldwide [1].

    CB banking has risen to the forefront of cell banking through the establishment of standardized recruiting, collection, processing, storage, product release, and quality control. CB transplantation is successfully used to treat myeloid leukemia, lymphatic leukemia, lymphoma, myelodysplasia, aplastic anemia, hemoglobinopathies, thalassemia, metabolic storage diseases, immune deficiency, autoimmune diseases, and other diseases in pediatric and adult patients [9–13]. In addition, CB banks may be public (donated CBUs used for unrelated patients in need) or private (CBUs saved for the use of the donating family), which has led to a public health debate. In this chapter, the current state of cell banking for clinical use will be discussed, and also recent advances in the generation, characterization, and bioprocessing of stem cells sourced from tissues other than CB—human pluripotent stem cells and induced pluripotent stem cells—for cell banking and clinical applications.

    II. Allogeneic and Autologous Cell Banking

    Public banks store allogeneic CBUs that have been donated by parents to be made available to registered transplant centers to provide CBUs to unrelated individuals [5]. Some banks may have provisions that a sibling or close relative may access related units for families with a predisposition to a particular genetic disease [5]. These mixed banks offer separate storage for CBUs that can be accessed through public registries and for others that are stored for purely private use [14]. Public CB banks do not charge for the donation, have a limited geographic collection region, and may be supported by government funds, grants, or philanthropy [15]. In addition, a fee is charged to the transplant patient’s insurance when a CBU is accessed for transplant [15].

    Private CB banks accept CB from families and store it on their behalf for possible autologous (for the donor only) or allogeneic (for a family member) transplantation; furthermore, the family is charged a fee for collection and processing (∼$1500–$2000 US) on acceptance of the units and an annual storage fee (∼$90–$200 US) that supports the operation of these private banks [15]. Consequently, these CBUs cannot be traced through the registries and are unavailable to the general public [16]. Based on the incidence of various pathologies, the possibility that a transplant will become necessary, and the probability of finding a compatible donor, it is estimated that the likelihood of privately stored CBUs being used for autologous purposes falls between 1:2500 (0.04%) and 1:20,000 (0.005%) [17,18]. The value of storing CBUs, therefore, may be realized by researchers who evaluate CB cells for pathologies other than hematological disorders such as heart disease, diabetes, stroke, traumatic brain and spinal cord injuries, and cancer [19–23]. Controversial issues in public CB banking include collection methods, processing techniques, and thawing methods [15]. CB can be collected in utero before delivery of the placenta by the delivering physician or midwife, or ex utero, which is a less invasive and more controlled technique, but is more expensive and requires additional trained personnel [15].

    III. Recruitment

    The legitimacy of public cell banking will depend on a number of factors. Public authorities are advised to take specific measures to ensure that sufficient donations are collected from different ethnic groups with different HLA patterns so that any patient needing a transplantation will be able to find an appropriate donor [24]. Moreover, a greater understanding of the impact of ethnicity on CB banking is required, as several recent studies have shown that ethnicity is associated with a higher risk of failing to meet banking criteria, lower CB volume, reduced total nucleated cell count, fewer CD34+ cells, and reduced colony-forming units [25]. Another important issue is that of informed consent for both allogeneic and autologous cell banking [26].

    IV. Regulations and Determination of Regulatory Guidance

    Regulatory oversight of therapeutics derived from biological sources has existed for some time. Due to the unique source of these products, assessment by traditional regulatory systems based on pharmaceutical quality-control parameters is difficult [27]. Growth within the field of regenerative medicine has led to increased use of novel transplantation therapies to repair or replace dysfunctional tissues and organs, which has led to increased public-health concerns. Consequently, established regulatory agencies throughout the world have developed new regulations for these products with the primary goal of minimizing infectious-disease risk. Regulators face challenges, as they must balance their roles as independent assessors with the needs of the overall public-health structure. Thus, regulators have recognized the need to assess life-saving therapies through systems that consider the risk–benefit ratios and include mechanisms for transparent and accountable release of products when full compliance with traditional manufacturing concepts is not possible [27].

    In the United States, products composed of human cells, tissues, and cellular and tissue-based products (HCT/Ps) are regulated by two divisions within the US Food and Drug Administration (FDA), the Center for Devices and Radiological Health (CDRH) for medical devices, and the Center for Biologics Evaluation and Research (CBER) (Table 1). Title 21 of the Code of Federal Regulations (CFR) comprises FDA rules, and the most current version of CFR 21 can be accessed directly from the FDA’s Web site or from the website of the Government Printing Office [28,29]. According to the FDA Regulation of HCT/Ps Product List, CBER regulates HCT/Ps under 21 CFR 1271.3(d)(1) and Section 361 of the Public Health Service (PHS) Act; furthermore, these HCT/Ps are regulated solely as 361 products when they meet all of the criteria in 21 CFR 1271.10(a):

    • Minimally manipulated;

    • Intended for a homologous use only as reflected by the labeling, advertising, or other indications of the manufacturer’s objective intent;

    • Not combined with another article, (except for water, crystalloids, or a sterilizing, preserving, or storage agent, if the addition of the agent does not raise new clinical safety concerns with respect to the HCT/P); and

    • Either:

    • Do not have a systemic effect and are not dependent upon the metabolic activity of living cells for their primary function; or

    • Have a systemic effect or are dependent upon the metabolic activity of the other cells for their primary function, and:

    • Are for autologous use;

    • Are for allogeneic use in a first- or second-degree relative; or

    • Are for reproductive use.

    HCT/Ps not covered by this compliance program or for which only certain provisions apply are discussed further on the FDA Web site [28]. Human somatic cell therapy and gene therapy products are also regulated by CBER under Section 351 of the PHS Act and/or the Food, Drug, and Cosmetic (FD&C) Act. This grouping includes products that the FDA has determined do not meet all of the criteria in 21 CFR 1271.10(a) and are regulated as drugs and/or biological products, refer to Table 1. Devices composed of human tissues are regulated by CDRH under the FD&C Act and device regulations, refer to Table 1. A section is also included for combination products, refer to Table 1.

    Table 1

    FDA Regulation of Human Cells, Tissues, and Cellular and Tissue-Based Products (HCT/Ps) Product List http://www.fda.gov/BiologicsBloodVaccines/TissueTissueProducts/RegulationofTissues

    21 CFR: Code of Federal Regulations Title 21; CBER: Center for Biologics Evaluation and Research; CDRH: Center for Devices and Radiological Health; FDA: US Food and Drug Administration; FD&C Act: Federal Food, Drug, and Cosmetic Act; HCT/Ps: Human Cells, Tissues, and Cellular and Tissue-Based Products; PHS Act: Public Health Service Act.

    V. Registration and Donor Eligibility Screening/Testing

    Within five days of engaging in the manufacture of an HCT/P, a facility must register with and submit to the FDA a list of each human-tissue product manufactured unless excepted by 21 CFR 1271.15. Manufacturing facilities must determine donor eligibility through appropriate screening and testing of HCT/P donors for risk factors for, and clinical evidence of, relevant communicable disease agents and diseases; as well as communicable disease risks associated with xenotransplantation. These procedures must be designed to ensure compliance with the requirements of subpart C, 21 CFR part 1271. Title 21 CFR part 1270 applies only to certain human tissue intended for transplantation (musculoskeletal, skin, and ocular), recovered before May 25, 2005, and requires donor screening and testing for only certain diseases (HIV, hepatitis B, and hepatitis C). Title 21 CFR part 1271, subpart C, applies to donors of additional cells and tissues, recovered on or after May 25, 2005, and require screening and testing of these donors for additional relevant communicable diseases. For example, 21 CFR part 1271, subpart C, applies to donors of hematopoietic stem/progenitor cells derived from peripheral and UCB, reproductive cells and tissue, human dura mater, and human heart valves, in addition to donors of musculoskeletal, skin, and ocular tissue. Title 21 CFR part 1271 also applies to HCT/Ps regulated as drugs, devices, or biological products, whereas 21 CFR part 1270, does not [28,29].

    VI. Current Good Tissue Practice (cGTP)

    To prevent the introduction, transmission, or spread of communicable diseases by HCT/Ps, manufacturing facilities must follow current good tissue practice (cGTP) requirements, which are the requirements in 21 CFR part 1271, subparts C and D, that govern the methods used in, and the facilities and controls used for, the manufacture of HCT/Ps, including but not limited to all steps in recovery, donor screening, donor testing, processing, storage, labeling, packing, and distribution (21 CFR 1271.150(a)). The core cGTP requirements as referenced in 21 CFR 1271.150(b) include requirements relating to:

    • Facilities (21 CFR 1271.190(a–b))

    • Environmental controls (21 CFR 1271.195(a))

    • Equipment (21 CFR 1271.200(a))

    • Supplies and reagents (21 CFR 1271.210(a–b))

    • Recovery (21 CFR 1271.215)

    • Processing and process controls (21 CFR 1271.220)

    • Labeling controls (21 CFR 1271.250(a–b))

    • Storage (21 CFR 1271.260(a–d))

    • Receipt, predistribution shipment, and distribution of an HCT/P (21 CFR 1271.265(a–d)).

    • Donor eligibility determinations, donor screening, and donor testing (21 CFR 1271.50, 1271.75, 1271.80,

    Enjoying the preview?
    Page 1 of 1