SlideShare a Scribd company logo
1 of 427
Download to read offline
DRUGS AND THE PHARMACEUTICAL SCIENCES
A Series of Textbooks and Monographs
1. Pharmacokinetics, Milo Gibaldi and Donald Perrier
2. Good Manufacturing Practices for Pharmaceuticals: A Plan for Total
Quality Control, Sidney H. Willig, Murray M. Tuckerman,
and William S. Hitchings IV
3. Microencapsulation, edited by J. R. Nixon
4. Drug Metabolism: Chemical and Biochemical Aspects, Bernard Testa
and Peter Jenner
5. New Drugs: Discovery and Development, edited by Alan A. Rubin
6. Sustained and Controlled Release Drug Delivery Systems, edited by
Joseph R. Robinson
7. Modern Pharmaceutics, edited by Gilbert S. Banker
and Christopher T. Rhodes
8. Prescription Drugs in Short Supply: Case Histories, Michael A. Schwartz
9. Activated Charcoal: Antidotal and Other Medical Uses, David O. Cooney
10. Concepts in Drug Metabolism (in two parts), edited by Peter Jenner
and Bernard Testa
11. Pharmaceutical Analysis: Modern Methods (in two parts), edited by
James W. Munson
12. Techniques of Solubilization of Drugs, edited by Samuel H. Yalkowsky
13. Orphan Drugs, edited by Fred E. Karch
14. Novel Drug Delivery Systems: Fundamentals, Developmental Concepts,
Biomedical Assessments, Yie W. Chien
15. Pharmacokinetics: Second Edition, Revised and Expanded, Milo Gibaldi
and Donald Perrier
16. Good Manufacturing Practices for Pharmaceuticals: A Plan for Total
Quality Control, Second Edition, Revised and Expanded, Sidney H. Willig,
Murray M. Tuckerman, and William S. Hitchings IV
17. Formulation of Veterinary Dosage Forms, edited by Jack Blodinger
18. Dermatological Formulations: Percutaneous Absorption, Brian W. Barry
19. The Clinical Research Process in the Pharmaceutical Industry, edited by
Gary M. Matoren
20. Microencapsulation and Related Drug Processes, Patrick B. Deasy
21. Drugs and Nutrients: The Interactive Effects, edited by Daphne A. Roe
and T. Colin Campbell
22. Biotechnology of Industrial Antibiotics, Erick J. Vandamme
23. Pharmaceutical Process Validation, edited by Bernard T. Loftus
and Robert A. Nash
24. Anticancer and Interferon Agents: Synthesis and Properties, edited by
Raphael M. Ottenbrite and George B. Butler
25. Pharmaceutical Statistics: Practical and Clinical Applications,
Sanford Bolton
26. Drug Dynamics for Analytical, Clinical, and Biological Chemists,
Benjamin J. Gudzinowicz, Burrows T. Younkin, Jr.,
and Michael J. Gudzinowicz
27. Modern Analysis of Antibiotics, edited by Adjoran Aszalos
28. Solubility and Related Properties, Kenneth C. James
29. Controlled Drug Delivery: Fundamentals and Applications, Second Edition,
Revised and Expanded, edited by Joseph R. Robinson and Vincent H. Lee
30. New Drug Approval Process: Clinical and Regulatory Management,
edited by Richard A. Guarino
31. Transdermal Controlled Systemic Medications, edited by Yie W. Chien
32. Drug Delivery Devices: Fundamentals and Applications, edited by
Praveen Tyle
33. Pharmacokinetics: Regulatory • Industrial • Academic Perspectives,
edited by Peter G. Welling and Francis L. S. Tse
34. Clinical Drug Trials and Tribulations, edited by Allen E. Cato
35. Transdermal Drug Delivery: Developmental Issues and Research
Initiatives, edited by Jonathan Hadgraft and Richard H. Guy
36. Aqueous Polymeric Coatings for Pharmaceutical Dosage Forms,
edited by James W. McGinity
37. Pharmaceutical Pelletization Technology, edited by Isaac Ghebre-Sellassie
38. Good Laboratory Practice Regulations, edited by Allen F. Hirsch
39. Nasal Systemic Drug Delivery, Yie W. Chien, Kenneth S. E. Su,
and Shyi-Feu Chang
40. Modern Pharmaceutics: Second Edition, Revised and Expanded,
edited by Gilbert S. Banker and Christopher T. Rhodes
41. Specialized Drug Delivery Systems: Manufacturing and Production
Technology, edited by Praveen Tyle
42. Topical Drug Delivery Formulations, edited by David W. Osborne
and Anton H. Amann
43. Drug Stability: Principles and Practices, Jens T. Carstensen
44. Pharmaceutical Statistics: Practical and Clinical Applications,
Second Edition, Revised and Expanded, Sanford Bolton
45. Biodegradable Polymers as Drug Delivery Systems, edited by
Mark Chasin and Robert Langer
46. Preclinical Drug Disposition: A Laboratory Handbook, Francis L. S. Tse
and James J. Jaffe
47. HPLC in the Pharmaceutical Industry, edited by Godwin W. Fong
and Stanley K. Lam
48. Pharmaceutical Bioequivalence, edited by Peter G. Welling,
Francis L. S. Tse, and Shrikant V. Dinghe
49. Pharmaceutical Dissolution Testing, Umesh V. Banakar
50. Novel Drug Delivery Systems: Second Edition, Revised and Expanded,
Yie W. Chien
51. Managing the Clinical Drug Development Process, David M. Cocchetto
and Ronald V. Nardi
52. Good Manufacturing Practices for Pharmaceuticals: A Plan for Total
Quality Control, Third Edition, edited by Sidney H. Willig
and James R. Stoker
53. Prodrugs: Topical and Ocular Drug Delivery, edited by Kenneth B. Sloan
54. Pharmaceutical Inhalation Aerosol Technology, edited by
Anthony J. Hickey
55. Radiopharmaceuticals: Chemistry and Pharmacology, edited by
Adrian D. Nunn
56. New Drug Approval Process: Second Edition, Revised and Expanded,
edited by Richard A. Guarino
57. Pharmaceutical Process Validation: Second Edition, Revised
and Expanded, edited by Ira R. Berry and Robert A. Nash
58. Ophthalmic Drug Delivery Systems, edited by Ashim K. Mitra
59. Pharmaceutical Skin Penetration Enhancement, edited by
Kenneth A. Walters and Jonathan Hadgraft
60. Colonic Drug Absorption and Metabolism, edited by Peter R. Bieck
61. Pharmaceutical Particulate Carriers: Therapeutic Applications, edited by
Alain Rolland
62. Drug Permeation Enhancement: Theory and Applications, edited by
Dean S. Hsieh
63. Glycopeptide Antibiotics, edited by Ramakrishnan Nagarajan
64. Achieving Sterility in Medical and Pharmaceutical Products, Nigel A. Halls
65. Multiparticulate Oral Drug Delivery, edited by Isaac Ghebre-Sellassie
66. Colloidal Drug Delivery Systems, edited by Jörg Kreuter
67. Pharmacokinetics: Regulatory • Industrial • Academic Perspectives,
Second Edition, edited by Peter G. Welling and Francis L. S. Tse
68. Drug Stability: Principles and Practices, Second Edition, Revised
and Expanded, Jens T. Carstensen
69. Good Laboratory Practice Regulations: Second Edition, Revised
and Expanded, edited by Sandy Weinberg
70. Physical Characterization of Pharmaceutical Solids, edited by
Harry G. Brittain
71. Pharmaceutical Powder Compaction Technology, edited by
Göran Alderborn and Christer Nyström
72. Modern Pharmaceutics: Third Edition, Revised and Expanded, edited by
Gilbert S. Banker and Christopher T. Rhodes
73. Microencapsulation: Methods and Industrial Applications, edited by
Simon Benita
74. Oral Mucosal Drug Delivery, edited by Michael J. Rathbone
75. Clinical Research in Pharmaceutical Development, edited by Barry Bleidt
and Michael Montagne
76. The Drug Development Process: Increasing Efficiency and Cost
Effectiveness, edited by Peter G. Welling, Louis Lasagna,
and Umesh V. Banakar
77. Microparticulate Systems for the Delivery of Proteins and Vaccines,
edited by Smadar Cohen and Howard Bernstein
78. Good Manufacturing Practices for Pharmaceuticals: A Plan for Total
Quality Control, Fourth Edition, Revised and Expanded, Sidney H. Willig
and James R. Stoker
79. Aqueous Polymeric Coatings for Pharmaceutical Dosage Forms:
Second Edition, Revised and Expanded, edited by James W. McGinity
80. Pharmaceutical Statistics: Practical and Clinical Applications,
Third Edition, Sanford Bolton
81. Handbook of Pharmaceutical Granulation Technology, edited by
Dilip M. Parikh
82. Biotechnology of Antibiotics: Second Edition, Revised and Expanded,
edited by William R. Strohl
83. Mechanisms of Transdermal Drug Delivery, edited by Russell O. Potts
and Richard H. Guy
84. Pharmaceutical Enzymes, edited by Albert Lauwers and Simon Scharpé
85. Development of Biopharmaceutical Parenteral Dosage Forms, edited by
John A. Bontempo
86. Pharmaceutical Project Management, edited by Tony Kennedy
87. Drug Products for Clinical Trials: An International Guide to Formulation •
Production • Quality Control, edited by Donald C. Monkhouse
and Christopher T. Rhodes
88. Development and Formulation of Veterinary Dosage Forms:
Second Edition, Revised and Expanded, edited by Gregory E. Hardee
and J. Desmond Baggot
89. Receptor-Based Drug Design, edited by Paul Leff
90. Automation and Validation of Information in Pharmaceutical Processing,
edited by Joseph F. deSpautz
91. Dermal Absorption and Toxicity Assessment, edited by Michael S. Roberts
and Kenneth A. Walters
92. Pharmaceutical Experimental Design, Gareth A. Lewis, Didier Mathieu,
and Roger Phan-Tan-Luu
93. Preparing for FDA Pre-Approval Inspections, edited by Martin D. Hynes III
94. Pharmaceutical Excipients: Characterization by IR, Raman, and NMR
Spectroscopy, David E. Bugay and W. Paul Findlay
95. Polymorphism in Pharmaceutical Solids, edited by Harry G. Brittain
96. Freeze-Drying/Lyophilization of Pharmaceutical and Biological Products,
edited by Louis Rey and Joan C. May
97. Percutaneous Absorption: Drugs–Cosmetics–Mechanisms–Methodology,
Third Edition, Revised and Expanded, edited by Robert L. Bronaugh
and Howard I. Maibach
98. Bioadhesive Drug Delivery Systems: Fundamentals, Novel Approaches,
and Development, edited by Edith Mathiowitz, Donald E. Chickering III,
and Claus-Michael Lehr
99. Protein Formulation and Delivery, edited by Eugene J. McNally
100. New Drug Approval Process: Third Edition, The Global Challenge,
edited by Richard A. Guarino
101. Peptide and Protein Drug Analysis, edited by Ronald E. Reid
102. Transport Processes in Pharmaceutical Systems, edited by
Gordon L. Amidon, Ping I. Lee, and Elizabeth M. Topp
103. Excipient Toxicity and Safety, edited by Myra L. Weiner
and Lois A. Kotkoskie
104. The Clinical Audit in Pharmaceutical Development, edited by
Michael R. Hamrell
105. Pharmaceutical Emulsions and Suspensions, edited by Francoise Nielloud
and Gilberte Marti-Mestres
106. Oral Drug Absorption: Prediction and Assessment, edited by
Jennifer B. Dressman and Hans Lennernäs
107. Drug Stability: Principles and Practices, Third Edition, Revised
and Expanded, edited by Jens T. Carstensen and C. T. Rhodes
108. Containment in the Pharmaceutical Industry, edited by James P. Wood
109. Good Manufacturing Practices for Pharmaceuticals: A Plan for Total
Quality Control from Manufacturer to Consumer, Fifth Edition, Revised
and Expanded, Sidney H. Willig
110. Advanced Pharmaceutical Solids, Jens T. Carstensen
111. Endotoxins: Pyrogens, LAL Testing, and Depyrogenation, Second Edition,
Revised and Expanded, Kevin L. Williams
112. Pharmaceutical Process Engineering, Anthony J. Hickey
and David Ganderton
113. Pharmacogenomics, edited by Werner Kalow, Urs A. Meyer
and Rachel F. Tyndale
114. Handbook of Drug Screening, edited by Ramakrishna Seethala
and Prabhavathi B. Fernandes
115. Drug Targeting Technology: Physical • Chemical • Biological Methods,
edited by Hans Schreier
116. Drug–Drug Interactions, edited by A. David Rodrigues
117. Handbook of Pharmaceutical Analysis, edited by Lena Ohannesian
and Anthony J. Streeter
118. Pharmaceutical Process Scale-Up, edited by Michael Levin
119. Dermatological and Transdermal Formulations, edited by
Kenneth A. Walters
120. Clinical Drug Trials and Tribulations: Second Edition, Revised
and Expanded, edited by Allen Cato, Lynda Sutton, and Allen Cato III
121. Modern Pharmaceutics: Fourth Edition, Revised and Expanded, edited by
Gilbert S. Banker and Christopher T. Rhodes
122. Surfactants and Polymers in Drug Delivery, Martin Malmsten
123. Transdermal Drug Delivery: Second Edition, Revised and Expanded,
edited by Richard H. Guy and Jonathan Hadgraft
124. Good Laboratory Practice Regulations: Second Edition, Revised
and Expanded, edited by Sandy Weinberg
125. Parenteral Quality Control: Sterility, Pyrogen, Particulate, and Package
Integrity Testing: Third Edition, Revised and Expanded, Michael J. Akers,
Daniel S. Larrimore, and Dana Morton Guazzo
126. Modified-Release Drug Delivery Technology, edited by
Michael J. Rathbone, Jonathan Hadgraft, and Michael S. Roberts
127. Simulation for Designing Clinical Trials: A Pharmacokinetic-
Pharmacodynamic Modeling Perspective, edited by Hui C. Kimko
and Stephen B. Duffull
128. Affinity Capillary Electrophoresis in Pharmaceutics and Biopharmaceutics,
edited by Reinhard H. H. Neubert and Hans-Hermann Rüttinger
129. Pharmaceutical Process Validation: An International Third Edition,
Revised and Expanded, edited by Robert A. Nash and Alfred H. Wachter
130. Ophthalmic Drug Delivery Systems: Second Edition, Revised
and Expanded, edited by Ashim K. Mitra
131. Pharmaceutical Gene Delivery Systems, edited by Alain Rolland
and Sean M. Sullivan
132. Biomarkers in Clinical Drug Development, edited by John C. Bloom
and Robert A. Dean
133. Pharmaceutical Extrusion Technology, edited by Isaac Ghebre-Sellassie
and Charles Martin
134. Pharmaceutical Inhalation Aerosol Technology: Second Edition,
Revised and Expanded, edited by Anthony J. Hickey
135. Pharmaceutical Statistics: Practical and Clinical Applications,
Fourth Edition, Sanford Bolton and Charles Bon
136. Compliance Handbook for Pharmaceuticals, Medical Devices,
and Biologics, edited by Carmen Medina
137. Freeze-Drying/Lyophilization of Pharmaceutical and Biological Products:
Second Edition, Revised and Expanded, edited by Louis Rey
and Joan C. May
138. Supercritical Fluid Technology for Drug Product Development, edited by
Peter York, Uday B. Kompella, and Boris Y. Shekunov
139. New Drug Approval Process: Fourth Edition, Accelerating Global
Registrations, edited by Richard A. Guarino
140. Microbial Contamination Control in Parenteral Manufacturing, edited by
Kevin L. Williams
141. New Drug Development: Regulatory Paradigms for Clinical Pharmacology
and Biopharmaceutics, edited by Chandrahas G. Sahajwalla
142. Microbial Contamination Control in the Pharmaceutical Industry, edited by
Luis Jimenez
143. Generic Drug Product Development: Solid Oral Dosage Forms, edited by
Leon Shargel and Izzy Kanfer
144. Introduction to the Pharmaceutical Regulatory Process, edited by
Ira R. Berry
145. Drug Delivery to the Oral Cavity: Molecules to Market, edited by
Tapash K. Ghosh and William R. Pfister
146. Good Design Practices for GMP Pharmaceutical Facilities, edited by
Andrew Signore and Terry Jacobs
147. Drug Products for Clinical Trials, Second Edition, edited by Donald
Monkhouse, Charles Carney, and Jim Clark
148. Polymeric Drug Delivery Systems, edited by Glen S. Kwon
149. Injectable Dispersed Systems: Formulation, Processing, and Performance,
edited by Diane J. Burgess
150. Laboratory Auditing for Quality and Regulatory Compliance,
Donald Singer, Raluca-Ioana Stefan, and Jacobus van Staden
151. Active Pharmaceutical Ingredients: Development, Manufacturing,
and Regulation, edited by Stanley Nusim
152. Preclinical Drug Development, edited by Mark C. Rogge and David R. Taft
153. Pharmaceutical Stress Testing: Predicting Drug Degradation, edited by
Steven W. Baertschi
154. Handbook of Pharmaceutical Granulation Technology: Second Edition,
edited by Dilip M. Parikh
155. Percutaneous Absorption: Drugs–Cosmetics–Mechanisms–Methodology,
Fourth Edition, edited by Robert L. Bronaugh and Howard I. Maibach
156. Pharmacogenomics: Second Edition, edited by Werner Kalow,
Urs A. Meyer and Rachel F. Tyndale
157. Pharmaceutical Process Scale-Up, Second Edition, edited by
Michael Levin
158. Microencapsulation: Methods and Industrial Applications, Second Edition,
edited by Simon Benita
159. Nanoparticle Technology for Drug Delivery, edited by Ram B. Gupta
and Uday B. Kompella
Nanoparticle Technology
for Drug Delivery
edited by
Ram B. Gupta
Auburn University
Auburn, Alabama
Uday B. Kompella
University of Nebraska Medical Center
Omaha, Nebraska
New York London
Published in 2006 by
Taylor & Francis Group
270 Madison Avenue
New York, NY 10016
© 2006 by Taylor & Francis Group, LLC
No claim to original U.S. Government works
Printed in the United States of America on acid-free paper
10 9 8 7 6 5 4 3 2 1
International Standard Book Number-10: 1-57444-857-9 (Hardcover)
International Standard Book Number-13: 978-1-57444-857-3 (Hardcover)
This book contains information obtained from authentic and highly regarded sources. Reprinted material is
quoted with permission, and sources are indicated. A wide variety of references are listed. Reasonable efforts
have been made to publish reliable data and information, but the author and the publisher cannot assume
responsibility for the validity of all materials or for the consequences of their use.
No part of this book may be reprinted, reproduced, transmitted, or utilized in any form by any electronic,
mechanical, or other means, now known or hereafter invented, including photocopying, microfilming, and
recording, or in any information storage or retrieval system, without written permission from the publishers.
For permission to photocopy or use material electronically from this work, please access www.copyright.com
(http://www.copyright.com/) or contact the Copyright Clearance Center, Inc. (CCC) 222 Rosewood Drive,
Danvers, MA 01923, 978-750-8400. CCC is a not-for-profit organization that provides licenses and registration
for a variety of users. For organizations that have been granted a photocopy license by the CCC, a separate
system of payment has been arranged.
Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and are used only
for identification and explanation without intent to infringe.
Library of Congress Cataloging-in-Publication Data
Catalog record is available from the Library of Congress
Visit the Taylor & Francis Web site at
http://www.taylorandfrancis.com
Taylor & Francis Group
is the Academic Division of Informa plc.
Preface
Products of nanotechnology are expected to revolutionize
modern medicine, as evidenced by recent scientific advances
and global initiatives to support nanotechnology and nano-
medicine research. The field of drug delivery is a direct bene-
ficiary of these advancements. Due to their versatility in
targeting tissues, accessing deep molecular targets, and con-
trolling drug release, nanoparticles are helping address chal-
lenges to face the delivery of modern, as well as conventional
drugs. Since the majority of drug products employ solids,
nanoparticles are expected to have a broad impact on drug
product development. The purpose of this book is to present
practical issues in the manufacturing and biological applica-
tion of nanoparticles. Drug delivery scientists in industry,
academia, and regulatory agencies, as well as students in bio-
medical engineering, chemical engineering, pharmaceutical
sciences, and other sciences with an interest in drug delivery,
iii
will find this book useful. It can also be used as a textbook for
drug delivery courses focusing on nanoparticles.
This book is organized into four sections. The first section
describes the distinguishing fundamental properties of nano-
particles (Chap. 1) as well as technologies for nanoparticle
manufacturing (Chaps. 2–4). Nanoparticles can be manufac-
tured by either breaking macro-particles using technologies
such as milling and homogenization (Chap. 2) or by building
particles from molecules dissolved in a solution using super-
critical fluid technology (Chap. 3). Nanoparticle manufacturing
and properties can be further optimized by employing poly-
mers or proteins as stabilizers (Chap. 4).
The second section describes the characterization of nano-
particles at the material or physicochemical level (Chap. 5) and
relates these properties to the delivery and effectiveness of
nanoparticles (Chap. 6) as well as toxicological characteristics
(Chap. 7).
The third section presents the various applications of
nanoparticles in drug delivery. Depending on the route and
purpose of drug delivery, the requirements for nanoparticulate
systems can vary. These aspects are discussed in Chapter 8 for
injectable delivery, Chapter 9 for oral delivery, Chapter 10 for
brain delivery, Chapter 11 for ocular delivery, and Chapter 12
for gene delivery.
Finally, the fourth section provides an overview of the
clinical, ethical, and regulatory issues of nanoparticle-based
drug delivery. These are evolving areas and the drug product
development experience with nanoparticles is limited. As
more data is gathered on the safety and efficacy of nanoparti-
culate systems, a clearer view will emerge.
Preparation of this book would not have been possible
without the valuable contributions from various experts in
the field. We deeply appreciate their timely contributions.
Also, we are thankful to our colleagues at Auburn University
and the University of Nebraska Medical Center for their
support in preparing this book.
Ram B. Gupta
Uday B. Kompella
iv Preface
Contents
Preface . . . . iii
Contributors . . . . xi
PART I: TECHNOLOGIES FOR NANOPARTICLE
MANUFACTURING
1. Fundamentals of Drug Nanoparticles . . . . . . . . 1
Ram B. Gupta
Introduction . . . . 1
Nanoparticle Size . . . . 2
Nanoparticle Surface . . . . 3
Nanoparticle Suspension and Settling . . . . 4
Magnetic and Optical Properties . . . . 6
Production of Nanoparticles . . . . 6
Biological Transport of Nanoparticles . . . . 12
Conclusions . . . . 17
References . . . . 18
v
2. Manufacturing of Nanoparticles by Milling
and Homogenization Techniques . . . . . . . . . . . 21
Rainer H. Mu€ller, Jan Mo€schwitzer, and
Faris Nadiem Bushrab
Introduction . . . . 21
Pearl/Ball-Milling Technology for the Production of
Drug Nanocrystals . . . . 25
Drug Nanocrystals Produced by
High-Pressure Homogenization . . . . 28
Production of Drug Nanocrystal Compounds by
Spray-Drying . . . . 33
Production in Nonaqueous Liquids . . . . 35
Production in Hot-Melted Matrices . . . . 37
Pelletization Techniques . . . . 41
Direct Compress . . . . 45
References . . . . 47
3. Supercritical Fluid Technology for
Particle Engineering . . . . . . . . . . . . . . . . . . . . . 53
Ram B. Gupta
Introduction . . . . 53
Supercritical CO2 . . . . 54
Solubility in Supercritical CO2 . . . . 55
Rapid Expansion of Supercritical Solution for
Particle Formation . . . . 59
RESS with Solid Cosolvent for Nanoparticle
Formation . . . . 63
Supercritical Antisolvent Process for Particle
Formation . . . . 66
SAS with Enhanced Mass (EM) Transfer
(SAS-EM) Process for Nanoparticle
Formation . . . . 69
Fundamentals Governing Particle Formation with
RESS and SAS . . . . 70
Other Applications of SCFs for
Particle Engineering . . . . 74
Safety and Health Issues . . . . 78
Conclusions . . . . 78
References . . . . 79
vi Contents
4. Polymer or Protein Stabilized Nanoparticles
from Emulsions . . . . . . . . . . . . . . . . . . . . . . . . . 85
Ram B. Gupta
Introduction . . . . 85
Emulsification Solvent Evaporation Process . . . . 86
Emulsification . . . . 87
Nanoparticle Hardening . . . . 93
Residual Solvent and Emulsifier . . . . 97
Protein Stabilized Nanoparticles . . . . 98
Conclusions . . . . 99
References . . . . 101
PART II: NANOPARTICLE CHARACTERIZATION
AND PROPERTIES
5. Physical Characterization of Nanoparticles . . . 103
Roy J. Haskell
Introduction . . . . 103
Measurement of Size . . . . 105
Available Methods . . . . 109
In Vitro Release . . . . 119
Example: Particle Size . . . . 121
Conclusions . . . . 130
References . . . . 132
6. Nanoparticle Interface: An Important
Determinant in Nanoparticle-Mediated
Drug/Gene Delivery . . . . . . . . . . . . . . . . . . . . . . 139
Sanjeeb K. Sahoo and Vinod Labhasetwar
Introduction . . . . 139
Influence of Emulsifier on
Pharmaceutical
Properties of Nanoparticles . . . . 140
Implication on Cellular Uptake/Toxicity/Gene
Delivery . . . . 148
Biodistribution . . . . 153
Conclusions . . . . 154
References . . . . 154
Contents vii
7. Toxicological Characterization of Engineered
Nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . 161
Paul J. A. Borm and Roel P. F. Schins
Introduction . . . . 161
Inhalation of Particles . . . . 164
Effects of Nanoparticles . . . . 170
Screening Engineered NP for Toxicological
Hazards . . . . 178
Conclusion . . . . 187
References . . . . 188
PART III: DRUG DELIVERY APPLICATIONS OF
NANOPARTICLES
8. Injectable Nanoparticles for Efficient Drug
Delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 199
Barrett Rabinow and Mahesh V. Chaubal
Introduction: Medical Needs Addressable by
Nanoparticulate Drug Delivery . . . . 199
Types of Carriers . . . . 209
Coating Functionality . . . . 215
External Assistance in Targeting . . . . 215
Drugs Incorporated . . . . 216
Clinical Development . . . . 217
Conclusions . . . . 220
References . . . . 221
9. Polymeric Nanoparticles for Oral Drug
Delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 231
Vivekanand Bhardwaj and Majeti Naga Venkata Ravi
Kumar
Introduction . . . . 231
Physiology of GIT with Relevance to Particulate
Uptake . . . . 232
Particle Size and Surface Charge: Critical Factors in
Particle Absorption . . . . 236
Bioadhesion . . . . 237
Tracer Techniques . . . . 241
viii Contents
In Vitro and In Vivo Models . . . . 243
Nanoparticle Formulation . . . . 244
Applications . . . . 253
Future Directions . . . . 260
References . . . . 262
10. Brain Delivery by Nanoparticles . . . . . . . . . . . 273
Svetlana Gelperina
Introduction . . . . 273
Biodistribution Studies . . . . 276
Pharmacological Activity . . . . 289
Mechanisms of Drug Delivery to the Brain by
Means of Polymeric NP . . . . 296
Conclusions . . . . 309
References . . . . 311
11. Nanoparticles for Ocular Drug Delivery . . . . . 319
Aniruddha C. Amrite and Uday B. Kompella
Introduction . . . . 319
Disposition of Nanoparticles in the Eye . . . . 322
Ocular Drug Delivery Enhancement Using
Nanoparticles . . . . 336
Safety and Tolerability of Particulate
Systems . . . . 347
Conclusions . . . . 352
References . . . . 353
12. DNA Nanoparticle Gene Delivery Systems . . . 361
Moses O. Oyewumi and Kevin G. Rice
Gene Delivery Vectors . . . . 361
Polymers Used to Prepare DNA
Nanoparticles . . . . 363
Physical Properties of DNA Nanoparticles . . . . 365
Biodistribution and Trafficking of DNA
Nanoparticles . . . . 371
Conclusions . . . . 372
References . . . . 373
Contents ix
PART IV: CLINICAL, ETHICAL, AND
REGULATORY ISSUES
13. Nanotechnology and Nanoparticles: Clinical,
Ethical, and Regulatory Issues . . . . . . . . . . . . 381
Makena Hammond and Uday B. Kompella
Introduction . . . . 381
Clinical Aspects . . . . 382
Environmental, Social, and
Ethical Issues . . . . 385
Regulatory Challenges . . . . 388
Conclusions . . . . 392
References . . . . 393
Index . . . . 397
x Contents
Contributors
Aniruddha C. Amrite Department of Pharmaceutical
Sciences, University of Nebraska Medical Center, Omaha,
Nebraska, U.S.A.
Vivekanand Bhardwaj Department of Pharmaceutics, National
Institute of Pharmaceutical Education and Research, Punjab,
India
Paul J. A. Borm Centre of Expertise in Life Sciences,
Zuyd University, Heerlen, The Netherlands
Faris Nadiem Bushrab Department of Pharmaceutical
Technology, Biotechnology and Quality Management, Freie
Universita¨t, Berlin, Germany
Mahesh V. Chaubal BioPharma Solutions, Baxter Healthcare,
Round Lake, Illinois, U.S.A.
xi
Svetlana Gelperina Institute of Molecular Medicine, Moscow
Sechenov Medical Academy, Moscow, Russia
Ram B. Gupta Department of Chemical Engineering, Auburn
University, Auburn, Alabama, U.S.A.
Makena Hammond College of Pharmacy, University of
Nebraska Medical Center, Omaha, Nebraska, and Virginia State
University, Petersburg, Virginia, U.S.A.
Roy J. Haskell Pfizer Corporation, Michigan Pharmaceutical
Sciences, Kalamazoo, Michigan, U.S.A.
Uday B. Kompella Department of Pharmaceutical Sciences,
College of Pharmacy, University of Nebraska Medical Center,
Omaha, Nebraska, U.S.A.
Majeti Naga Venkata Ravi Kumar Department of
Pharmaceutics, National Institute of Pharmaceutical Education
and Research, Punjab, India
Vinod Labhasetwar Department of Pharmaceutical Sciences,
University of Nebraska Medical Center, Omaha, Nebraska, U.S.A.
Jan Mo¨schwitzer Department of Pharmaceutical Technology,
Biotechnology and Quality Management, Freie Universita¨t, Berlin,
Germany
Rainer H. Mu¨ ller Department of Pharmaceutical Technology,
Biotechnology and Quality Management, Freie Universita¨t, Berlin,
Germany
Moses O. Oyewumi Division of Medicinal and Natural Products
Chemistry, College of Pharmacy, University of Iowa, Iowa City,
Iowa, U.S.A.
Barrett Rabinow BioPharma Solutions, Baxter Healthcare,
Round Lake, Illinois, U.S.A.
Kevin G. Rice Division of Medicinal and Natural Products
Chemistry, College of Pharmacy, University of Iowa, Iowa City,
Iowa, U.S.A.
xii Contributors
Sanjeeb K. Sahoo Department of Pharmaceutical Sciences,
University of Nebraska Medical Center, Omaha, Nebraska, U.S.A.
Roel P. F. Schins Institut fur Umweltmedizinische Forschung
(IUF), University of Du¨sseldorf, Du¨sseldorf, Germany
Contributors xiii
1
Fundamentals of Drug Nanoparticles
RAM B. GUPTA
Department of Chemical Engineering,
Auburn University, Auburn, Alabama, U.S.A.
INTRODUCTION
In pharmaceutics, %90% of all medicines, the active ingredi-
ent is in the form of solid particles. With the development in
nanotechnology, it is now possible to produce drug nanoparti-
cles that can be utilized in a variety of innovative ways. New
drug delivery pathways can now be used that can increase
drug efficacy and reduce side effects. For example, in 2005,
the U.S. Food and Drug Administration approved intra-
venously administered 130-nm albumin nanoparticles loaded
with paclitaxel (AbraxaneTM
) for cancer therapy, which epito-
mizes the new products anticipated based on nanoparticulate
systems. The new albumin/paclitaxel–nanoparticle formula-
tion offers several advantages including elimination of
PART I: TECHNOLOGIES FOR
NANOPARTICLE MANUFACTURING
1
toxicity because of cremophor, a solvent used in the previous
formulation, and improved efficacy due to the greater dose of
the drug that can be administered and delivered. For better
development of the nanoparticulate systems, it is essential to
understand the pharmaceutically relevant properties of nano-
particles, which is the purpose of this chapter and this book
in general. In the following narrative, some fundamental prop-
erties of nanoparticles including their size, surface area, set-
tling velocity, magnetic and optical properties, and biological
transport are brought into the perspective of drug delivery.
NANOPARTICLE SIZE
To put the size of nanoparticles in perspective, Table 1 com-
pares sizes of various objects. Because of the comparable size
of the components in the human cells, nanoparticles are of
great interest in drug delivery. It appears that nature, in
making the biological systems, has extensively used nan-
ometer scale. If one has to go hand in hand with nature in
treating the diseases one needs to use the same scale, whether
it is correcting a faulty gene, killing leprosy bacteria sitting
inside the body cells, blocking the multiplication of viral gen-
ome, killing a cancer cell, repairing the cellular metabolism,
or preventing wrinkles or other signs of aging. One cannot
use a human arm to massage the hurt leg of an ant. Size
matching is important in carrying out any activity. Drug
delivery is aimed at influencing the biochemistry of the body.
Table 1 Typical Size of Various Objects
Object Size (nm)
Carbon atom 0.1
DNA double helix (diameter) 3
Ribosome 10
Virus 100
Bacterium 1,000
Red blood cell 5,000
Human hair (diameter) 50,000
Resolution of unaided human eyes 100,000
2 Gupta
The basic unit of the biological processes is the cell and the
biochemical reactions inside it. With the advent of nanoparti-
cles it is now possible to selectively influence the cellular pro-
cesses at their natural scales.
NANOPARTICLE SURFACE
We can generally see and discern objects as small as
100,000 nm (100 mm). It is only in the past 300 years, with the
invention of microscopes, that we can see smaller objects.
Today, one can see objects as small as individual atoms (about
0.1 nm) using the scanning probe microscope. Owing to their
small size, nanoparticles exhibit interesting properties, mak-
ing them suitable for a variety of drug delivery applications.
The number of molecules present on a particle surface
increases as the particle size decreases. For a spherical solid
particle of diameter d, surface area per unit mass,Sg, is given as
Sg ¼
pd2
4
 
pd3
rs
6
 À1
¼
3
2drs
ð1Þ
where rs is the solid density. If the molecular diameter is s,
then the percentage of molecules on the surface monolayer is
given as
%Surface molecules ¼
ð4=3Þp½d3
À ðd À sÞ3
Š
ð4=3Þp½d3Š
100
¼ 100
s
d
 3
À3
s
d
 2
þ3
s
d
 !
ð2Þ
For a typical low–molecular weight drug molecule
of 1-nm diameter, %surface molecules are calculated in
Table 2. It is interesting to see that for a 10,000-nm (or
10-mm) particle, a very small percentage of the molecules are
present on the surface. Hence, the dissolution rate is much
lower for the microparticles when compared to nanoparticles.
When the particles are of nanometer length scale, surface
irregularities can play an important role in adhesion, as the
irregularities may be of the same order as the particles (1).
Fundamentals of Drug Nanoparticles 3
Nanoparticles can show a strong adhesion because of the
increased contact area for van der Waals attraction. For
example, Lamprecht et al. (2) observed differential uptake/
adhesion of polystyrene particle to inflamed colonic mucosa,
with the deposition 5.2%, 9.1%, and 14.5% for 10-mm, 1000-nm,
and 100-nm particles, respectively.
NANOPARTICLE SUSPENSION AND SETTLING
Because of the small size of the nanoparticles, it is easy to
keep them suspended in a liquid. Large microparticles preci-
pitate out more easily because of gravitational force, whereas
the gravitational force is much smaller on a nanoparticle.
Particle settling velocity, v, is given by Stokes’ law as
v ¼
d2
gðrs À rlÞ
18ml
ð3Þ
where g is gravitation acceleration (9.8 m/sec at sea level), rl
is liquid density (997 kg/m3
for water at 25
C), ml is viscosity
(0.00089 Pa/sec for water at 25
C). For various particles sizes,
settling velocities are calculated in Table 3 for a solid density
(rs) of 1700 kg/m3
.
Thermal (Brownian) fluctuations resist the particle
settlement. According to Einstein’s fluctuation–dissipation
theory, average Brownian displacement x in time t is given as
x ¼
ffiffiffiffiffiffiffiffiffiffiffiffiffi
2kBTt
pmd
s
ð4Þ
Table 2 % Surface Molecules in Particles
Particle size (nm) Surface molecules (%)
1 100.00
10 27.10
100 2.97
1,000 0.30
10,000 0.03
4 Gupta
where kB is the Boltzman constant (1.38 Â 10À23
J/K), and T is
temperature in Kelvin. Table 4 shows displacements for
particles of varying sizes in water at 25
C. The Brownian
motion of a 1000-nm particle due to thermal fluctuation in
water is 1716 nm/sec, which is greater than the settling velocity
of 430 nm/sec. Hence, particles below 1000 nm in size will not
settle merely because of Brownian motion. This imparts an
important property to nanoparticles, that they can be easily
kept suspended despite high solid density. Large microparti-
cles easily settle out from suspension because of gravity, hence
such suspensions need to carry a ‘‘shake well before use’’ label.
Also, a microparticle suspension cannot be used for injection.
For the nanoparticles, the gravitational pull is not stronger
than the random thermal motion of the particles. Hence, nano-
paticle suspensions do not settle, which provides a long self-life.
However, settling can be induced using centrifugation if
needed for particle separation. Particle velocity under centri-
fugation is given as:
v ¼
p
9
xd2
ðrs À rlÞ
m
rpm
60
 2
ð5Þ
Table 4 Brownian Motion of the Particles
Particle size (nm) Brownian displacement (nm in 1 sec)
1 54,250
10 17,155
100 5,425
1,000 1,716
10,000 543
Table 3 Particle Settling Velocities
Particle size (nm) Settling velocity (nm/sec)
1 0.00043
10 0.043
100 4.30
1,000 430
10,000 43,005
Fundamentals of Drug Nanoparticles 5
where the centrifugal rotation is rotations per minute (rpm).
For various particle sizes, centrifugal velocities calculated
for a solid density (rs) of 1700 kg/m3
in water at 0.1 m from
the axis of rotation are presented in Table 5.
MAGNETIC AND OPTICAL PROPERTIES
Small nanoparticles also exhibit unique magnetic and optical
properties. For example, ferromagnetic materials become
superparamagnetic below about 20 nm, i.e., the particles do
not retain the magnetization because of the lack of magnetic
domains; however, they do experience force in the magnetic
field. Such materials are useful for targeted delivery of drugs
and heat. For example, interaction of electromagnetic pulses
with nanoparticles can be utilized for enhancement of drug
delivery in solid tumors (3). The particles can be attached to
antibodies directed against antigens in tumor vasculature
and selectively delivered to tumor blood vessel walls. The
local heating of the particles by pulsed electromagnetic radia-
tion results in perforation of tumor blood vessels, microcon-
vection in the interstitium, and perforation of cancer cell
membrane, and therefore provides enhanced delivery of drugs
from the blood into cancer cells with minimal thermal and
mechanical damage to the normal tissues.
Gold and silver nanoparticles show size-dependent opti-
cal properties (4). The intrinsic color of nanoparticles changes
with size because of surface plasmon resonance. Such nano-
particles are useful for molecular sensing, diagnostic, and
imaging applications. For example, gold nanoparticles can
exhibit different colors based on size (Table 6).
PRODUCTION OF NANOPARTICLES
Although any particle of a size 1-mm diameter is a nanopar-
ticle, several national initiatives are encouraging the develop-
ment of particles 100 nm as they might exhibit some unique
physical properties, and hence potentially different and use-
ful biological properties. However, achieving sizes 100 nm
6 Gupta
Table5CentrifugalVelocitiesoftheParticles
Centrifugalvelocity(m/sec)oftheparticles
fordifferentrotationspeeds
Particlesize(nm)1,000rpm2,000rpm5,000rpm20,000rpm50,000rpm
17.66Â10À12
3.06Â10À11
1.91Â10À10
3.06Â10À9
1.91Â10À8
107.66Â10À10
3.06Â10À9
1.91Â10À8
3.06Â10À7
1.91Â10À6
1007.66Â10À8
3.06Â10À7
1.91Â10À6
3.06Â10À5
1.91Â10À4
1,0007.66Â10À6
3.06Â10À5
1.91Â10À4
3.06Â10À3
1.91Â10À2
10,0007.66Â10À4
3.06Â10À3
1.91Â10À2
3.06Â10À1
1.91
Fundamentals of Drug Nanoparticles 7
is more readily feasible with hard materials compared to drug
and polymer molecules, which are soft materials. For hard
materials, such as silica, metal oxides, and diamonds with
melting points above 1000
C, nanoparticles in the 1–100 nm
size range have been prepared. However, for drugs that are
usually soft materials with melting point below 300
C parti-
cles in the 1–100 nm size range are more difficult to prepare.
For this reason, it is a reasonable goal to aim at 300 nm par-
ticles for drug and polymer materials. There are several suc-
cess stories for pharmaceutical materials in this size range.
Table 6 Size-Dependent Color Variation of Gold Nanoparticles
Wavelength for maximum absorption (nm)
Nanoparticle size (nm) In water In AOT/water/isooctane (w0 ¼ 10)
9 519 535
20 523 531
30 525 535
40 526 545
52 528 543
59 535 546
79 550 560
100 567 583
Abbreviation: AOT, sodium bis(2-ethyl hexyl)sulfosuccinate.
Source: From Ref. 5.
Table 7 Number of Molecules in a Spherical Particle
Particle diameter Particle volume (mL) Number of molecules
0.58 nm 8.18 Â 10À22
1
1 nm 4.19 Â 10À21
5.05
10 nm 4.19 Â 10À18
5.05 Â 103
100 nm 4.19 Â 10À15
5.05 Â 106
500 nm 5.24 Â 10À13
6.31 Â 108
1 mm 4.19 Â 10À12
5.05 Â 109
5 mm 5.24 Â 10À10
6.31 Â 1011
1 mm 4.19 Â 10À3
5.05 Â 1018
Note: Drug molecular weight ¼ 500 and solid density ¼ 1 g/mL.
8 Gupta
Production of nanoparticles of soft materials is much more
challenging than that of hard materials because of the high
stickiness of the former. The bulk pharmaceuticals are avail-
able in solids of large sizes (e.g., 1-mm-diameter powder), which
can be often easily solubilized in solvent to obtain molecular
size. Hence, there are two extremes of sizes: molecular size
(each particle containing one molecule) and large size (e.g., each
particle containing of the order of 1018
molecules). For a drug of
500 molecular weight and 1 g/mL solid density, the numbers of
molecules in different size particles are given in Table 7.
Hence, to obtain nanoparticles in the 50–300 nm range
for drug delivery, one requires of the order of 104
–108
mole-
cules in each particle. This size has to be achieved from either
solution phase (single molecule) or millimeter-size particle
(1018
molecules). The first approach is where the particle is
broken down to nano size, whereas in the second approach,
the particle will be built up from molecules. The two general
approaches for the production of drug nanoparticles are
sketched in Figure 1.
Milling of Large Particles or
Breaking-Down Process
Comminuting or grinding or milling is the oldest mechanical
unit operation for size reduction of solids and for producing
large quantities of particulate materials. Here, the material
is subjected to stress, which results in the breakage of the par-
ticle. Usually, the applied stress is more concentrated on the
Figure 1 Schematic of the two general nanoparticle production
techniques.
Fundamentals of Drug Nanoparticles 9
already present cracks in the material, which causes crack
propagation leading to fracture. With the decreasing particle
size, materials exhibit increasing plastic behavior making it
more difficult to break small particles than large particles.
For many materials, a limit in the grindability can be reached
where subject to further grinding, no decrease in the particle
size is observed (6). An empirical index, known as Bond work
index (Wi) has been developed, which represents energy
required for grinding (7).
Wi ¼ 10 d
À1=2
product À d
À1=2
feed
 
ð6Þ
To reduce the size of a 1-mm particle, the energy
required in terms of Bond index is given in Table 8 for various
product sizes.
Hence, it is very energy intensive to go down to nanopar-
ticles-size range. Other than size, parameters of importance
are: (i) toughness/brittleness (in tough materials, stress
causes plastic deformation, whereas in brittle materials
cracks are propagated; hence, size reduction of brittle materi-
als is easier than for tough materials; sometimes, a material
can be cooled to embrittle), and (ii) hardness, abrasiveness,
particle shape and structure, heat sensitivity (only about 2%
of the applied energy goes to size reduction, the rest is con-
verted to heat; hence, heat-sensitive drugs require cooling),
and explodability (most pharmaceuticals are organic materi-
als; as the size is reduced air combustibility of the material
increases, hence proper inerting is needed).
Table 8 Energy Need (Bond Work Index) for Reducing
Size of 1-mm-Diameter Particles
Product diameter (mm) Energy required (Bond work index)
100 0.68
10 2.85
1 9.68
0.5 13.83
0.1 31.31
10 Gupta
Most of the pharmaceutical size reduction operations uti-
lize high-shear wet milling for the production of nanoparticles.
Milling is explained in detail in chapter 2. Typical operation
time for the wet milling may be hours to days, hence the drug
has to show stability in that time period, otherwise milling
cannot be used for unstable drugs. In addition, one has to
be aware of contamination due to milling media.
Precipitation from Solution or
Building-Up Process
In this process, a drug is dissolved in a solvent to achieve
molecular solution. Then, the nanoparticle precipitate is
obtained either by removing the solvent rapidly or by mixing
an antisolvent (nonsolvent) to the solution, reducing its solu-
bilizing strength. Initially, nuclei are formed, which grow
because of condensation and coagulation giving the final par-
ticles. If the rate of desolubilization is slow, then sticky nuclei/
particles are formed that have a higher tendency of agglo-
meration, giving large-size final particles. For example, if a
drug is dissolved in a solvent (e.g., toluene) and then an
Figure 2 Variation of the particle size as the antisolvent and its
mixing are varied in the solvent–antisolvent precipitation process.
Fundamentals of Drug Nanoparticles 11
antisolvent (e.g., methanol) is added with mild mixing, one
will obtain drug precipitate of typically 1 mm particle size
(Fig. 2). To obtain nanoparticles, a high desolubilization rate
is needed, or use of a surfactant is required, which can
isolate the particles until they are completely dry. Based on
these requirements, two general methods for nanoparticle
production are available: (i) supercritical fluid process, and
(ii) emulsification–diffusion process. In the precipitation pro-
cess, one can add compounds (e.g., polymers for controlled
release) that will coprecipitate with the drug for smart drug
delivery applications.
The key aspect of getting nanoparticles of the desired
size and size distribution is to control both the rate of antisol-
vent action and the particle coagulation. Precipitation-based
processes are described in chapters 3 and 4.
BIOLOGICAL TRANSPORT OF NANOPARTICLES
For drug delivery, most of the sites are accessible through
either microcirculation by blood capillaries or pores present
at various surfaces and membranes. Most of the apertures,
openings, and gates at cellular or subcellular levels are of
nanometer size (Table 9); hence, nanoparticles are the most
suited to reach the subcellular level. One of the prime require-
ments of any delivery system is its ability to move around
freely in available avenues and by crossing various barriers
that may come in the way. Regarding the human body, the
major passages are the blood vessels through which materials
are transported in the body. The blood vessels are not left in
any organ as an open outlet of the pipe, rather they become
thinner and thinner and are finally converted to capillaries
through branching and narrowing. These capillaries go to
the close vicinity of the individual cells. After reaching their
thinnest sizes, the capillaries start merging with each other
to form the veins. These veins then take the contents back
to the heart for recirculation. Hence, the supply chain in the
body is not in the form of a pipe having an open inlet to the
organ and outlet away from the organ. Consequently, for
12 Gupta
any moiety to remain in the vasculature, it needs to have its
one dimension narrower than the cross-sectional diameter of
the narrowest capillaries, which is about 2000 nm. Actually,
for efficient transport the nanoparticle should be smaller
than 300 nm.
But, just moving in the vessels does not serve the drug
delivery purpose. The delivery system must reach the site
at the destination level. This requires crossing of the blood
capillary wall to reach the extracellular fluid of the tissue
and then again crossing of other cells, if they are in the
way, and entering the target cell. These are the major bar-
riers in the transit. A nanoparticle has to do a lot during this
sojourn of the carrier through the vessels (capillaries) and
across the barriers.
There are two routes for crossing the blood capillaries
and other cell layers, i.e., transcellular and paracellular. In
Table 9 Approximate Sizes of Components in a
Typical 20-mm Human Tissue Cell
Component Size (nm)
Ribosomes 25
Golgi vesicles 30–80
Secretary vesicles 100–1000
Glycogen granules 10–40
Lipid droplets 200–5000
Vaults 55
Lysosomes 500–1000
Proteasomes 11
Peroxisomes 500–1000
Mitochondria 500–1000
Superfine filaments 2–4
Microfilaments 5–7
Thick filaments 15
Microtubules 25
Centrioles 150
Nuclear pores 70–90
Nucleosomes 10
Chromatin 1.9
Source: From Refs. 8, 9.
Fundamentals of Drug Nanoparticles 13
the transcellular route, the particulate system has to enter
the cell from one side and exit the cell from the other side
to reach the tissue. The particulate system has to survive
the intracellular environment to reach the target tissue.
The other route is paracellular. In this, the particlulate
system is not required to enter the cell; instead, it moves
between the cells. These intercellular areas are known as
the junctions. Passing through the junctions would obviate
destruction of the carrier by the cell system. Paracellular
movement of moieties including ions, larger molecules, and
leukocytes is controlled by the cytoskeletal association of tight
junctions and the adherence junctions called apical junction
complex. While tight junctions act as a regulated barrier,
the adherence junctions are responsible for the development
and stabilization of the tight junctions. Different epithelial
and endothelial barriers have different permeabilities mainly
because of the differences in the structure and the presence of
tight junctions. While epithelia and brain capillary endothe-
lium exhibit a high degree of barrier function, the vascular
endothelium in other tissues has greater permeability. The
tight junctions control the paracellular transport. For exam-
ple, diffusion of large molecules may not be feasible, but
migration of white cells is allowed. Understanding of this reg-
ulation mechanism is important as this might enable us to
pave the way for the movement of nanoparticles in the body
without actually entering into the unintended cells.
As the nanoparticle-based drug delivery is achieved by
particle transport, it is important to understand the blood
flow rates and volumes of various organs and tissues. Consid-
ering the body’s distribution network, the blood vascular sys-
tem, the body could be divided into several compartments
based on the distributional sequencing and differentiation
by the blood vascular system (Table 10).
Nanoparticles can have deep access to the human body
because of the particle size and control of surface properties.
Experiments by Jani et al. (13,14) have elegantly demon-
strated the size effect. Polystyrene particles in the size range
50–3000 nm were fed to rats daily for 10 days at a dose of
1.25 mg/kg. The extent of absorption of the 50-nm particles
14 Gupta
Table10VolumesandBloodSupplyofDifferentBodyRegions
TissueVolume(mL)
Volumeofbloodin
equilibriumwithtissue(mL)
Bloodflow
(mL/min)(mL/dLinanorgan/min)
Viscera(wellperfused)
Adrenals2062100500
Kidneys3007651240410
Thyroid204980400
Graymatter75037160080
Heart30014824080
Othersmallglandsandorgans160508050
Liverwithportalsystem3,900979158041
Whitematter75010016021
Leantissues(moderatelyperfused)
Redmarrow1,400741209
Muscle30,0009803.25
Skin(nutritive)3,000983.25
Nonfatsubcutaneous4,80043701.5
Adiposetissues(poorlyperfused)
Fattymarrow2,20037602.7
Fat10,0001232002
Others
Bonecortex6,400%0%0%0
Arterialblood1,400
Venousblood4,000
Lungparenchymaltissue6001400
Airinlung2.5þhalfthe
tidalvolume
Note:Cardiacoutput,6480mL/min;dataareofstandardmen:age30–39years,bodyweight70kg,andsurfacearea1.83mÀ2
.
Source:FromRefs.10–12.
Fundamentals of Drug Nanoparticles 15
was 34% and that of the 100-nm particles was 26%. Of the
total absorption, about 7% (50 nm) and 4% (100 nm) were
accounted for in the liver, spleen, blood, and bone marrow.
Particles 100 nm did not reach the bone marrow, and those
300 nm were absent from the blood. Particles were absent in
the heart or the lung tissue. The rapid clearance of circulating
particles from the bloodstream coupled with their high uptake
by liver and spleen can be overcome by reducing the particle
size, and by making the particle surface hydrophilic with
coatings, such as poloxamers or poloxamines (15).
Gaur et al. (16) observed that 100-nm nanoparticles of
polyvinylpyrrolidone had a negligible uptake by the macro-
phages in liver and spleen, and 5–10% of these nanoparticles
remain in circulation even eight hours after intravenous
injection. Because of longer residence in the blood, nanoparti-
cles have potential therapeutic applications, particularly in
cancer; the cytotoxic agents encapsulated in these particles
can be targeted to tumors while minimizing the toxicity to
the reticuloendothelial system.
Desai et al. (17) studied the effect of poly(d,l-lactide-co-
glycolide) (PLGA). particle size (100 nm, 500 nm, 1 mm, and
10 mm) on uptake in rat gastrointestinal tissue. The uptake
of 100-nm-size particles by the intestinal tissue was 15–250-
fold higher compared to the larger-size microparticles. The
uptake also depends on the type of tissue (i.e., Peyer’s patch
and nonpatch) and the location (i.e., duodenum or ileum).
Depending on the particle size, Peyer’s patch tissue had a
2–200-fold higher uptake of particles than the nonpatch tis-
sue. The 100-nm particles were diffused throughout the
submucosal layers, while the larger-size particles were predo-
minantly localized in the epithelial lining of the tissue,
because of the microparticle exclusion phenomena in the
gastrointestinal mucosal tissue.
Hillyer and Albrecht (18) studied the gastrointestinal
uptake and subsequent tissue/organ distribution of 4-, 10-,
28-, and 58-nm-diameter metallic colloidal gold particles fol-
lowing oral administration to mice. It was found that colloidal
gold uptake is dependent on the particle size: smaller particles
cross the gastrointestinal tract more readily. Interestingly,
16 Gupta
they observed that the particle uptake occurs in the small
intestine by persorption through single, degrading enterocytes
in the process of being extruded from a villus.
Cellular uptake is greater for nanoparticles compared to
microparticles. In cultured human retinal pigment epithelial
cells, an increase in the mass uptake of particles was observed
with decreasing particle size in the range of 20–2000 nm poly-
styrene particles (19). Further, no saturable uptake was
observed for these particles up to a concentration of 500 mg/
mL. With 20-nm nanoparticles, the uptake by the 1-cm2
cell
monolayer was as high as $20%.
Because of possible differences in particle uptake, gene
expression efficiencies can also be improved with smaller par-
ticles. Prabha et al. (20) studied relative transfectivity of 70-
and 202-nm-PLGA nanoparticles in cell culture. The smaller
particles showed a 27-fold higher transfection than the larger
nanoparticles in COS-7 cell line and a fourfold higher trans-
fection in HEK-293 cell line.
CONCLUSIONS
Nanoparticles offer unique properties as compared to micro-
or macroparticles. Salient features include the following:
 Small size.
 High surface area.
 Easy to suspend in liquids.
 Deep access to cells and organelles.
 Variable optical and magnetic properties.
 Particles smaller than 200 nm can be easily sterilized
by filtration with a 0.22-mm filter.
Drugs, being mostly organic compounds, are more sticky
in nature as compared to inorganic materials, such as silica or
metal oxides. Hence, it is harder to make smaller nanoparti-
cles of drugs compared with hard materials. Drug nanoparti-
cles can be produced either by milling of macroparticles or by
fast precipitation from solutions, as described in the following
chapters.
Fundamentals of Drug Nanoparticles 17
REFERENCES
1. Chow TS. Size-dependent adhesion of nanoparticles on rough
substrates. J Phys: Condens Matter 2003; 15(suppl 2):L83–L87.
2. Lamprecht A, Schafer U, Lehr C-M. Size-dependent bioadhe-
sion of micro- and nanoparticulate carriers to the inflamed
colonic mucosa. Pharm Res 2001; 18(suppl 6):788–793.
3. Barros APH. Synthesis and Agglomeration of Gold Nanoparti-
cles in Reverse Micelles. M.S. thesis, University of Puerto Rico,
Mayaguez, PR, 2005.
4. Esenaliev RO. Radiation and nanoparticles for enhancement
of drug delivery in solid tumors. PCT Int Appl 2000, WO
2000002590.
5. Kelly L, Coronado E, Zhao LL, Schatz GC. The optical proper-
ties of metal nanoparticles: the influence of size, shape, and
dielectric environment. J Phys Chem B 2003; 107:668–677.
6. Prior M. Size reduction. In: Kirk-Othmer, Othmer DF, eds.
Encyclopedia of Chemical Technology. John Wiley  Sons,
Inc., 2000.
7. Bond FC. Crushing and grinding calculations. Can Min Metal
Trans 1954; 57:466–472.
8. Alberts B, Bray D, Lewis J, Raff M, Roberts K, Watson JD.
The Molecular Biology of the Cell. 2nd ed. New York: Garland
Publishing, Inc., 1989.
9. Benjamin L. Genes V. New York: Oxford University Press,
1995.
10. Oie S, Benet LZ. The effect of route of administration and
distribution on drug action. In: Banker GS, Rhodes CT, eds.
Modern Pharmaceutics. New York: Marcel Dekker, 1996.
11. Dedrick RL, Bischoff KB. Pharmacokinetics in applications of
the artificial kidney. Chem Eng Progr Symp Ser 1968; 64:32–44.
12. Mapleson WW. An electric analogue for uptake and exchange
of inert gases and other agents. J Appl Physiol 1963; 18:
197–204.
13. Jani P, Halbert GW, Langridge J, Florence AT. Nanoparticle
uptake by the rat gastrointestinal mucosa: quantitation and
18 Gupta
particle size dependency. J Pharm Pharmacol 1990; 42(suppl 12):
821–826.
14. Jani P, Halbert GW, Langridge J, Florence AT. The uptake and
translocation of latex nanospheres and microspheres after oral
administration to rats. J Pharm Pharmacol 1989; 41(suppl 12):
809–812.
15. Rudt S, Muller RH. In vitro phagocytosis assay of nano- and
microparticles by chemiluminescence. III. Uptake of differ-
ently sized surface-modified particles, and its correlation to
particles properties and in vivo distribution. Eur J Pharm
Sci 1993; 1:31–39.
16. Gaur U, Sahoo SK, De TK, Ghosh PC, Maitra A, Ghosh PK.
Biodistribution of fluoresceinated dextran using novel nano-
particles evading reticuloendothelial system. Int J Pharm
2000; 202(suppl 1–2):1–10.
17. Desai MP, Labhasetwar V, Amidon GL, Levy RJ. Gastrointest-
inal uptake of biodegradable microparticles: effect of particle
size. Pharm Res 1996; 13(suppl 12):1838–1845.
18. Hillyer, JF, Albrecht RM. Gastrointestinal persorption and tis-
sue distribution of differently sized colloidal gold nanoparti-
cles. J Pharm Sci 2001; 90(suppl 12):1927–1936.
19. Aukunuru JV, Kompella UB. In vitro delivery of nano- and
micro-particles to retinal pigment epithelial (RPE) cells. Drug
Deliv Technol 2002; 2(suppl 2):50–57.
20. Prabha S, Zhou W-Z, Panyam J, Labhasetwar V. Size-
dependency of nanoparticle-mediated gene transfection: stu-
dies with fractionated nanoparticles. Int J Pharm 2002;
244(suppl 1–2):105–115.
Fundamentals of Drug Nanoparticles 19
2
Manufacturing of Nanoparticles by
Milling and Homogenization
Techniques
RAINER H. MU¨ LLER, JAN MO¨ SCHWITZER, and
FARIS NADIEM BUSHRAB
Department of Pharmaceutical Technology,
Biotechnology and Quality Management,
Freie Universita¨t, Berlin, Germany
INTRODUCTION
The number of newly developed drugs having a poor solubility
and thus exhibiting bioavailability problems after oral admini-
stration is steadily increasing. Estimates by the pharma-
ceutical companies are that about 40% of the drugs in the
pipelines are poorly soluble, and as high as 60% of the com-
pounds come directly from the synthesis route (1). Therefore,
since a number of years the pharmaceutical development is
21
focused on formulation approaches to overcome solubility and
related bioavailability problems, so that these new compounds
are available for clinical use. Often forgotten, the problem of
poor solubility arises even before the preclinical phase, which
means that when screening new compounds for pharmaco-
logical activity a test formulation needs to be able to lead to
sufficiently high blood levels. Therefore, there is an urgent
need to come up with a smart formulation approach.
One has to differentiate between specific and nonspecific
formulations for increasing solubility and, subsequently, bio-
availability. Specific approaches can only be applied to certain
drug molecules, e.g., in case of cyclodextrins (CDs) to mole-
cules that fit into the respective CD ring. In the area of
CDs, research is focused on CD derivates with higher solubi-
lity of the CD itself and simultaneously reduced side effects of
these excipients; for example, the recent development of Cap-
tisol1
CDs (2,3). On the other hand, the nonspecific
formulation approaches are applicable to almost any drug
molecule (apart from a few exceptions). Such a nonspecific
formulation approach since many years is micronization,
which means converting relatively coarse drug particles to
micrometer crystals with a mean diameter in the range of
approximately 2–5 mm, and a corresponding size distribution
approximately between 0.1 and 20 mm (4). Here, the increase
in the surface area leads to an increase in the dissolution velo-
city. That means micronization is a formulation approach to
overcome the bioavailability problems of drugs of the biophar-
maceutical specification class II (BSC II). Drugs of class II are
sufficiently permeable but the rate limiting step is a too low
dissolution velocity (i.e., low solubility in general is correlated
with low dissolution velocity, law by Noyes–Whitney). Now-
adays however, many of the new compounds are so poorly
soluble that micronization is not sufficient to overcome a too
low oral bioavailability. Consequently, the next step taken
was to move from micronization to nanonization.
By going down one more dimension from the microrange
to the nanoworld there is a distinct increase in the surface area
and related dissolution velocity. For example, when moving
from a spherical 50 mm particle to micronized 5 mm particles,
22 Mu€ller et al.
the total surface area enlarges by a factor of 10, moving to
500 nm nanocrystals by a factor of 100. However, there is an
additional—but often forgotten—effect further increasing the
dissolution velocity, that is, the increase in saturation solubi-
lity cs when moving to sizes below 1 mm. Because of the strong
curvature of the particles, they possess an increased dissolu-
tion pressure comparable to the increased vapor pressure of
ultrafine aerosol droplets. The theoretical background is pro-
vided by the Kelvin equation and the Ostwald–Freundlich
equation, which will not be discussed here in detail (5). Accord-
ing to the Noyes–Whitney equation the dissolution velocity
dc/dt is proportional to the concentration gradient cs À cx (cs—
saturation solubility, cx—concentration in surrounding med-
ium, bulk concentration). The increases in saturation solubility
of nanocrystals reported are by a factor of about 2 to 4–6 [(6,7)
and unpublished data]. The increase is even more pronounced
when the nanosized drug material is not crystalline but amor-
phous. Preparation of amorphous oleanolic acid nanoparticles
increased the saturation solubility up to 10-fold in relation to
the coarse drug powder (8). Nanonization has the advantage
that it practically can be applied to more or less any drug
material. In general, even highly water-sensitive drugs can
be reduced to drug nanocrystals, even stored in the form of
an aqueous nanosuspension (drug nanocrystals dispersed in
aqueous surfactant/stabilizer medium). For example, aqueous
Paclitaxel nanosuspension proved to be stable over a period
of four years stored at 4
C, i.e., more than 99% of the drug
was recovered intact (9). On the other hand, aqueous Paclitaxel
solution degrades to an extent of 80% within 25 minutes (10).
Drug nanocrystals can be produced by bottom-up or top-
down technologies. In the case of bottom-up technologies, one
starts with the molecules in solution and moves via associ-
ation of these molecules to the formation of solid particles,
i.e., it is a classical precipitation process (11). To our know-
ledge, there is presently no pharmaceutical product on the
market based on precipitation technology. There are a num-
ber of reasons, discussed in detail in Ref. (12). Briefly, the
use of solvents creates additional costs. In addition, a prere-
quisite for precipitation is that the drug is at least soluble
Manufacturing of Nanoparticles 23
in one solvent, and this solvent needs to be miscible with a
nonsolvent. Many of the newly developed compounds; how-
ever, are poorly soluble in aqueous and simultaneously in
nonaqueous media, thus excluding this formulation approach.
In the case of top-down technologies, one starts with a coarse
material and applies forces to disintegrate into the nanosize
range. The diminution technologies can be categorized into
two principal classes:
1. Pearl/ball milling.
2. High-pressure homogenization, and other processes.
There are two products on the market based on the
pearl/ball-millingtechnologybythecompanyNANOSYSTEMS
E´ LAN (13). Rapamune1
coated tablet is the more convenient
formulation for the patient compared to the drug solution
(Rapamune solution). The bioavailability of the tablet is
27% higher than the solution form (14). Rapamune1
was
introduced in the market in 2002 by the company Wyeth.
The second is Emend1
, introduced in the market in 2003 by
the company MSD1
, Sharp  Dohme Gmbh. It is a capsule
composed of sucrose, microcrystalline cellulose (MCC), hypro-
lose, and sodium dodecylsulfate (SDS) (15).
Also, the products based on drug nanocrystals produced
with high-pressure homogenization are in clinical phases.
Therefore, these two technologies are reviewed in this chapter
because of their relevance for the pharmaceutical market.
Drug nanocrystals are of high relevance to pharmaceuti-
cal products; therefore, it is not surprising that most of the
research and development are being done in pharmaceutical
companies, especially looking at the production process itself.
Of course as a consequence, articles published by companies
are very low in number to protect internal knowledge; primar-
ily, only published patents are accessible. Even less literature
is available on how to transfer the drug nanosuspensions to
the final products, i.e., tablets, capsules, and pellets for
oral administration or aqueous/lyophilized nanosuspensions
for intravenous injection. Producing drug nanocrystals is
relatively easy compared to the much more sophisticated
technology to formulate a final drug dosage form. A final
24 Mu€ller et al.
traditional drug dosage form has to be based on patient conve-
nience. However, to fully benefit from the special properties
of nanocrystals, they need to be released as ultrafine, non-
aggregated suspension from the final dosage form. It could
be shown that in the case of strong nanocrsytal aggregation,
the dissolution velocity is reduced (16). Therefore, the tricky
business is how to transfer the drug nanosuspension to
dosage forms with optimized release properties. This chapter
also describes the production of tablets, capsules, and pellets.
PEARL/BALL-MILLING TECHNOLOGY FOR THE
PRODUCTION OF DRUG NANOCRYSTALS
Traditional equipment used for micronization of drug
powders such as rotor–stator colloid mills (Netzsch) or jet
mills (Retsch) are of limited use for the production of nano-
crystals. For example, jet milling leads to a drug powder with
a size range of roughly between 0.1 and 20 mm, containing
only a very small fraction of about 10% in the nanometer
range (4). However, it could be shown when running a pearl
mill over a sufficiently long milling time, that drug nanosus-
pensions can be obtained (13,17,18). These mills consist of a
milling container filled with fine milling pearls or larger-sized
balls. The container can be static and the milling material is
moved by a stirrer; alternatively, the complete container is
moved in a complex movement leading consequently to move-
ment of the milling pearls.
There are different milling materials available, tradi-
tionally steel, glass, and zircon oxide are used. New materials
are special polymers, i.e., hard polystyrene. A problem asso-
ciated with the pearl milling technology is the erosion from
the milling material during the milling process. Buchmann
et al. (19) reported about the formation of glass microparti-
cles when using glass as milling material. In general, very
few data have been published on contamination of pharma-
ceutical drug nanosuspensions by erosion from the milling
material. Most data have been given in the discussions
after oral presentations; figures from such discussions range
Manufacturing of Nanoparticles 25
from 0.1 to 70 ppm contamination. Of course it should be
noted that the extent of erosion depends on the solid con-
centration of the macrosuspension to be processed, the
hardness of the drug, and based on this, the required milling
time and milling material. Apart from the milling material,
the erosion from the container also needs to be considered.
For the parts in contact with the product various materials
are offered by companies producing pearl mills depending
on the area of application, such as technical purposes,
food, or the pharmaceutical industry. Normally, product con-
tainers are made of steel and can be covered with various
materials to fulfill the required quality specifications of the
formulation.
Surfactants or stabilizers have to be added for the
physical stability of the produced nanosuspensions. In the
production process the coarse drug powder is dispersed by
high-speed stirring in a surfactant/stabilizer solution to yield
a macrosuspension. The choice of surfactants and stabilizers
depends not only on the properties of the particles to be
suspended (e.g., affinity of surfactant/stabilizer to the crystal
surface) but also on the physical principles (electrostatic vs.
steric stabilization) and the route of administration. In gen-
eral, steric stabilization is recommended as the first choice
because it is less susceptible to electrolytes in the gut or
blood. Electrolytes reduce the zeta potential and subse-
quently impair the physical stability, especially of ionic sur-
factants. In many cases an optimal approach is the
combination of a steric stabilizer with an ionic surfactant,
i.e., the combination of steric and electrostatic stabilization.
There is a wide choice of various charged surfactants in case
of drug nanocrystals for oral administration. Even relatively
‘‘nasty’’ surfactants, such as the membrane damaging SDS,
can be used, of course within the concentration accepted
for oral administration, e.g., the formulation of Emend (15).
SDS as a low molecular weight surfactant diffuses fast to
particle surfaces; it has excellent dispersion properties.
Adsorption onto the particle surface leads to high zeta poten-
tial values providing good physical stabilities. In case of par-
enteral drug nanocrystals, the choice is limited; e.g., for
26 Mu€ller et al.
intravenous injection, accepted are lecithins, Poloxamer 188,
Tween1
80, low molecular weight polyvinylpyrrolidone
(PVP), sodium glycocholate (in combination with lecithin).
Drug nanocrystal suspensions for parenteral administrations
need to be sterile, depending on the administration route and
the volume they need to be pyrogene-free. Production of par-
enteral drug nanosuspension using pearl mills is much more
tedious compared to producing oral drug nanosuspensions.
The equipment needs to be sterilized and the product needs
to be separated from the milling pearls by a preferentially
aseptic separation process. A terminal sterilization by auto-
claving is only possible with a number of products (20).
The use of an ionic stabilizer such as lecithin is recom-
mended when autoclaving nanosuspensions. The autoclaving
temperature of 121
C leads to dehydration of steric stabili-
zers, which reduces their ability to stabilize the suspensions.
Gamma irradiation of nanosuspensions is an alternative, but
is less favoured by the pharmaceutical industry due to regu-
latory requirements (e.g., proof of absence of toxic radicals,
etc.). From the industrial point of view, in many cases a
well-documented aseptic production is easier for the produc-
tion of formulations for parenteral administration than
gamma irradiation.
There are a number of pearl mills available on the
market, ranging from laboratory-scale to industrial-scale
volumes. The ability for large-scale production is an essential
prerequisite for the introduction of a product to the market.
One advantage of the pearl mills, apart from being low-cost
products, is their ability for scaling up. Assuming, for reasons
of simplicity, hexagonal packaging of the milling pearls, 76%
of the milling chamber volume will be filled by the pearls. In
case of a 1000 L mill this corresponds to 760 L milling mate-
rial; based on the apparent density of zircon oxide pearls
being 3.69 kg/L, this corresponds to 2.8 tons of milling mate-
rial. Figure 1 shows the solution for this problem, a pearl mill
with an external suspension container. The suspension is con-
tinuously pumped through the pearl mill. This approach
reduces the weight of the pearl mill itself, but it prolongs
the milling times.
Manufacturing of Nanoparticles 27
DRUG NANOCRYSTALS PRODUCED BY
HIGH-PRESSURE HOMOGENIZATION
Theoretical Aspects
High-pressure homogenization is a technology that has been
applied for many years in various areas for the production of
emulsions and suspensions. A distinct advantage of this
technology is its ease for scaling up, even to very large
volumes. High-pressure homogenization is currently used
in the food industry, e.g., homogenization of milk. In the
Figure 1 DISPERMAT1
SL: schematic view of a bead mill using
recirculation method. Source: From Ref. 21.
28 Mu€ller et al.
pharmaceutical industry parenteral emulsions are produced
by this technology. Commercial products such as Intralipid1
and Lipofundin1
possess a mean droplet diameter in the
range of 200–400 nm (photon correlation spectroscopy data)
(22). In the mid-1990s of the last century drug nanosuspen-
sions produced with high-pressure homogenization were
developed (23–27). Typical pressures for the production of
drug nanosuspensions are 1000–1500 bar (corresponding to
100–150 Mpa, 14504–21756 psi); the number of required
homogenization cycles vary from 10 to 20 depending on the
properties of the drug. Most of the homogenizers used are
based on the piston-gap principle, an alternative is the jet-
stream technology (Fig. 2).
The Microfluidizer1
(MicrofluidicsTM
Inc., U.S.A.) is
based on the jet-stream principle. Two streams of liquid col-
lide, diminution of droplets or crystals is achieved mainly by
particle collision, but occurrence of cavitation is also consid-
ered. The Microfluidizer has also been described for the pro-
duction of drug nanosuspensions; however, according to the
patent 10–50 cycle passes were required (28). Such a high
Figure 2 Basic homogenization principles: piston-gap (left) and
jet-stream arrangement (right). In the piston-gap homogenizer the
macrosuspension coming from the sample container is forced to
pass through a tiny gap (e.g., 10 mm), particle diminution is affected
by shear force, cavitation, and impaction. In jet-stream homogeni-
zers the collision of two high-velocity streams leads to particle
diminution mainly by impact forces.
Manufacturing of Nanoparticles 29
cycle number is not convenient for the production scale. Based
on our own experiences, the Microfluidizer can be used for the
production of drug nanosuspensions in the case of soft drugs.
In the case of harder drugs, a larger fraction of particles in
the micrometer range remain, which do not exhibit the
increase in saturation solubility because of their too large size.
For many years cavitation was considered as the major
force leading to particle diminution in the high-pressure
homogenization process. Consequently, most high-pressure
homogenization patents in various application areas focus
on water as a dispersion medium. In the piston-gap homoge-
nizer the liquid is forced through a tiny homogenization
gap, typically in the size range of 5–20 mm (depending on
the pressure applied and the viscosity of the dispersion med-
ium). Using a Micron Lab 40 the suspension is supplied from
a metal cylinder by a piston, the cylinder diameter is approxi-
mately 3 cm. The suspension is moved by the piston having an
applied pressure between 100 and 1500 bar. In principle the
piston-gap homogenizer corresponds to a tube system in
which the tube diameter narrows from 3 to 5–20 mm. Accor-
ding to the Bernoulli equation, the streaming velocity and
dynamic pressure increase extremely, the static pressure in
the gap falls below the vapor pressure of water at room tem-
perature. A liquid boils when its vapor pressure is equal to the
static pressure, which means water starts boiling in the gap
at room temperature leading to the formation of gas bubbles.
The formation of gas bubbles leads to pressure waves disrup-
ting oil droplets or disintegrating crystals. When leaving the
homogenization gap, the static pressure increases to normal
air pressure, which means the water does not boil any more
and the gas bubbles collapse. Collapsing of the gas bubbles
(implosion) leads again to shock waves contributing to
diminution. There are different definitions of cavitation in
the literature, describing cavitation either as the formation
of gas bubbles in high streaming liquids or as the formation
and subsequent implosion of these gas bubbles.
At the end of the 1990s it was found that similar efficient
particle diminution can be achieved by homogenization in
nonaqueous media such as oils and liquid polyethylene glycols
30 Mu€ller et al.
(PEGs), which means media with low vapor pressure. In the
case of low vapor pressure liquids, the cavitation in the homo-
genization gap is distinctly reduced or does not exist at all.
Figure 3 shows the change in static pressure when homoge-
nizing in water as dispersion medium (left) and in a low-vapor
liquid, whereas the static pressure does not fall below the
vapor pressure (right).
Based on the aforementioned, cavitation does not seem to
be essential for a diminution effect. Major forces are droplet or
particle collision and the shear forces occurring in this highly
turbulent fluid in the gap possessing a high kinetic energy.
Homogenization in nonaqueous liquids has advantages for cer-
tain pharmaceutical final dosage forms. Preparation of drug
nanocrystals in PEGs or oils (e.g., Miglyol 812 or 829) leads
to nanosuspensions that can directly be filled into capsules
(see the following) (29,30). It is also possible to homogenize in
melted nonaqueous matrices, which are solid at room tempera-
ture. Solidification of such a matrix leads to a fixation of drug
nanocrystals in the solid matrix, thus minimizing or avoiding
crystal contact and subsequent crystal fusion/growth.
Figure 3 Variation of the static pressure (—) within the homoge-
nizing gap. In the case of water the static pressure falls below the
vapor pressure (left), whereas in the case of low-vapor media (right)
the static pressure stays above the vapor pressure.
Manufacturing of Nanoparticles 31
As a consequent next step, after homogenization in water
(100% water) and homogenization in nonaqueous media
(0% water), homogenization was performed in mixtures con-
taining different percentages of water (1–99% water). The dis-
persion media were water mixed with water-miscible liquids
(e.g., alcohols, glycerol). Preparation of drug nanosuspensions
in water–ethanol mixtures is favorable for producing dry pro-
ducts, because later the spray drying can be performed under
milder conditions when using such a mixture. Homogeniza-
tion in water–glycerol mixtures (2.25% of water-free glycerol)
leads to isotonic drug nanosuspensions for parenteral
administration.
The laboratory scale homogenizers used by our group are
the continuous and batch Micron Lab 40 (APV Systems,
Unna, Germany). In the batch version, the batch size is a
minimum of 20 mL and a maximum of 40 mL. A minimum
of 20 mL is required for the machine to maintain the homoge-
nization pressure because smaller volumes cannot be pro-
cessed. In the batch Micron Lab 40, the homogenizer is
equipped with two product containers having a maximum
volume of 1000 mL. Considering the dead volume in the
machine, a minimum batch size of about 200 mL is recom-
mended. The advantage of the batch Micron Lab 40 is the
relatively small batch volume, but unfortunately it is not pro-
duced any more. The successor model by the company APV is
the APV-1000; however, the minimum batch size for this
homogenizer is 150 mL (31). Scaling up to a size suitable for
the production of clinical batches was performed using a
Lab 60 unit. This homogenizer has a homogenization capacity
of 60 L/hr, and can be qualified and validated (32). The com-
mercially available Lab 60 was modified by equipping it with
10 L double-walled product containers; processing is possible
in a continuous loop mode (2 kg batch) or alternatively in a
batch mode (5–10 kg batch). Because of the termination of
the production of the Lab 60 an APV 1000 or APV 2000 is
recommended for a batch size in this range. Larger-scale
machines from APV are the Gaulin 5.5 (160 L homogenization
capacity per hour) or the Rannie 55 (600 L homogenization
capacity per hour) at a pressure of 1500 bar.
32 Mu€ller et al.
Alternative suppliers of piston-gap homogenizers are the
companies Avestin1
(33) and GEA Niro Soavi (34).
PRODUCTION OF DRUG NANOCRYSTAL
COMPOUNDS BY SPRAY-DRYING
For the production of tablets, an aqueous nanosuspension can
be used as granulation fluid or a dry form of the nanosuspen-
sion, powder, or granulate can be employed. Starting from an
aqueous macrosuspension containing the original coarse
drug powder, surfactant, and water-soluble excipient, the
homogenization process can be performed in an easy one step
yielding a fine aqueous nanosuspension. In a subsequent step
the water has to be removed from the suspension to obtain a
dry powder. One method of removing the water from the for-
mulation is freeze drying, but it is complex and cost-intensive
leading to a highly sensitive product (35,36). Another simple
and most suitable method for the industrial production is
spray drying. The drug nanosuspension can directly be pro-
duced by high-pressure homogenization in aqueous solutions
of water-soluble matrix materials, e.g., polymers [PVP, poly-
vinylalcohol or long chained PEG, sugars (saccharose, lac-
tose), or sugar alcohols (mannitol, sorbitol)]. Afterward the
aqueous drug nanosuspension can be spray dried under ade-
quate conditions; the resulting dry powder is composed of
drug nanocrystals embedded in a water-soluble matrix (37).
Figure 4 schematically represents the whole production pro-
cess of drug nanocrystal-loaded spray-dried compounds.
The loading capacity of the solid powder with drug nano-
crystals can be adjusted by varying the concentrations of exci-
pient and surfactant in the original aqueous nanosuspension.
One aim of a solid nanoparticulate system is releasing the
drug nanocrystals after administration in the gastrointestinal
tract (GI) as a fine nonaggregated suspension; the other is to
increase the physical stability for long-term storage. Contact
of the drug nanocrystals is averted by fixation within the
matrix. Thereby, the probability of physical instabilities as,
e.g., aggregation and ripening are in principle clearly avoided
Manufacturing of Nanoparticles 33
or minimized to a negligible extent. However, appropriate
investigations have shown a relation between the loading
capacity of the compounds and the releasing behavior, as well
as the storage stability. Exceeding a certain maximum load-
ing capacity of the matrix with drug nanocrystals has an
increasing negative effect on particle crystal growth and on
release as fine dispersion (38).
Figure 5 shows the volume distribution of two spray-
dried formulations A and B with increasing drug nanocrystal
loadings after release in water. The formulation with the
highest drug concentration, formulation B, clearly shows a
negligible low but detectable aggregated volume fraction
Figure 4 Two-step process of the production of drug nanocrystal-
loaded compounds: the drug nanosuspension obtained by high-
pressure homogenization (Micron Lab 40) is further processed by
spray drying using a Mini Bu¨chi. Drug nanocrystals embedded in
the matrix are obtained.
34 Mu€ller et al.
and consequently, a reduced percentage within the lower
nanometer range.
PRODUCTION IN NONAQUEOUS LIQUIDS
To avoid the removal of water after high-pressure homogeni-
zation in aqueous media, homogenization can be performed
directly in nonaqueous media. A number of nonaqueous
media are suitable as dispersion media for drug nanocrystals.
For example, PEG and triglycerides or self-emulsifying drug
delivery systems are ideal liquid candidates and are suited
Figure 5 LD volume distributions of spray-dried formulations of
Tween1
80 stabilized amphotericin B nanocrystals in PVP matrix
after redispersion in water. An adequate volume of distilled water
(22–23
C) was added to the compounds to obtain a 1% drug nano-
suspension after release from the matrix. LD measurements were
started after gently stirring until the matrix material was comple-
tely dissolved. (ÁÁÁÁ): Formulation A: 15.4% AmphoB, 7.7% Tween
80, 76.9% PVP (Kollidon 25); (—): formulation B: 50.0% AmphoB,
25.0% Tween 80, 25.0% PVP (Kollidon 25); (–): original nanosus-
pension, 1% AmB, 0.5% Tween 80. Abbreviation: LD, laser diffrac-
tion; PVP, polyvinylpyrrolidone.
Manufacturing of Nanoparticles 35
for direct filling of hard or soft gelatine capsules (39). The pro-
duction process can be easily performed similar to the process
in water.
Influence of the Dispersion Media
Forces caused by shearing, cavitation, and impaction are domi-
nant for the diminution of drug particles during the homoge-
nizing process. However, the physical properties of the
dispersion media/suspension as well as the type of homogeni-
zer and geometry of its dissipation zone highly influence these
forces and consequently, the diminution. Especially, the visc-
osity of the suspension shows significant effect on the proper-
ties of the homogenized products.
According to the law by Hagen–Poiseuille, flow rate, pres-
sure, tube diameter, as well as the viscosity of the streaming
suspension are interdependent in laminar flow systems. The
Micron Lab 40 works with a constant flow rate. According to
the input requirements the homogenization pressure is auto-
matically adjusted by the width of the homogenizing gap (40).
In this correlation the viscosity of the fluid/suspension can be
considered as a determining factor for the width of the gap.
Using a Micron Lab 40 simplified a doubling of the width is
observed when decoupling the viscosity. Thus, increasing visc-
osities alter the flow conditions within the homogenizing
region. A decrease in flow velocity and an increase in gap
volume clearly influence the homogenization results. Figure 6
shows the calculated width of the homogenization gap of the
Micron Lab 40 in dependency on the fluid viscosity.
Broadening of the homogenization gap leads to decreased
shear forces and kinetic energies of the nanocrystals; con-
sequently, the lower forces affect the breaking of the particles.
In summary, the grade of particle diminution is deter-
mined by the forces acting on each drug particle during the
homogenization process and the drug properties (e.g., hard-
ness of crystal, number of imperfections in crystal, and per-
centage of amorphous fraction) (41). A particle breaks if
the acting force is higher than the breakage stress. The max-
imal dispersivity of a nanosuspension is reached if further
36 Mu€ller et al.
homogenization cycles show no more effect on particle size
distribution. In this case, the acting forces are not high enough
for inducing particle breakage and further diminution.
For the production of drug nanosuspensions in nonaque-
ous media the maximal dispersivity has to be investigated for
each drug and dispersion medium. In principle, the media
with considerably higher viscosities than water require a
higher number of homogenization cycles to achieve identical
or similar particle sizes and distributions to lower-viscosity
media. Figure 7 shows the particle size distribution of Ampho-
tericin B nanoparticles, using laser diffraction (LD), after
high-pressure homogenization in different dispersion media.
PRODUCTION IN HOT-MELTED MATRICES
A further possibility for the production of drug nanocrystals
in solid matrices is high-pressure homogenization in hot
Figure 6 Width of the homogenizing gap as a function of the kine-
matic viscosity of the fluid (Micron Lab 40) at a homogenization
pressure of 1500 bar.
Manufacturing of Nanoparticles 37
melts. It offers advantages over production in aqueous solu-
tion and subsequent spray drying. The process is completely
anhydrous, avoiding possible drug degradation or instabi-
lities. The production can directly be performed by hot high-
pressure homogenization in melted material (38,42). The
homogenizers Micron Lab 40, batch and continuous, were
equipped with temperature control jackets placed around
the sample/product containers. Working temperatures up to
100
C (heated with water) or higher (heated with silicon oil)
can be selected depending on the melting temperature of
the used matrix material. For batch operation, solidification
has to be averted between each homogenizing cycle. For
homogenizers working in the continuous mode, the product
containers must be also heated. Figure 8 shows the tempera-
ture control devices for the continuous and batch versions of
the Micron Lab 40.
As the first production step, a presuspension has to be
formed consisting of a melted matrix with the addition of
the drug powder and surfactant. In the following production
Figure 7 LD particle diameters 25–90% of Amphotericin-B nano-
crystals after high-pressure homogenization in PEGs of various
chain lengths and thus viscosities. Abbreviations: LD, laser diffrac-
tion; PEG, polyethylene glycol.
38 Mu€ller et al.
step, the hot presuspension can be directly homogenized in
the temperature-controlled homogenizer. After reaching the
envisaged particle size and size distribution, the suspension
can be solidified at room temperature by applying controlled
cooling. Figure 9 shows the principle production process of
drug nanocrystals in hot melts.
Subsequently, the solid nanodispersions obtained can be
processed to granulates by milling, for filling capsules or
tablet compaction. Alternatively, the hot melt can directly
be filled into hard gelatine or hydroxypropyl methylcellulose
(HPMC) capsules (Fig. 10).
The absence of water during the whole production pro-
cess as well as the short processing times and the one-step
process to the final product are especially to be noted using
the hot melt method. Given these advantages, this techno-
logy also has limitations, which have to be compared with
the other technologies (e.g., homogenization in water) for
the production of solid nanosuspensions. High-pressure
homogenization—identical to pearl milling—can only be
performed up to a certain drug concentration. Suspensions
Figure 8 Micron Lab 40 with temperature control jackets (J):
temperature control jacket for the discontinuous Micron Lab 40
(left) around the sample container (S) containing the suspension
to be homogenized, for continuous Micron Lab 40 (right) with jack-
ets around the sample container and additional jackets around the
two 1000 mL product containers (P).
Manufacturing of Nanoparticles 39
Figure 9 Schematic of the process utilizing melted matrices: the
coarse drug material is added to the solid matrix material, which
is then melted for dispersing the drug. The nanosuspension is
obtained by high-pressure homogenization. Subsequent cooling
leads to drug nanocrystals embedded in a solid matrix.
Figure 10 Capsules filled with granulated PEG 2000 containing
Amphotericin-B nanocrystals (left), tablets produced by compaction
of the granulate (right). Abbreviation: PEG, polyethylene glycol.
40 Mu€ller et al.
can show paste-like properties at high solids content (e.g.,
30% or 40%). The resulting rheological properties, espe-
cially high viscosity, lead to suboptimal flow conditions
within the homogenization gap. Depending on the homogeni-
zer design, some suspensions with higher viscosities can also
be processed (e.g., feeding the suspension to the homogenizer
by applying air pressure or using a piston, e.g., PANDA
range of GEA, Niro-Soavi, Stansted homogenizers) (43). For
example, using a Stansted homogenizer a suspension of
40% solid can be processed without any problem (44). How-
ever, it should be noted that the viscosity of a suspension
does not only depend on the solid content but also on the
size, size distribution, and shape of the particles. Depending
on these factors particles can form three-dimensional struc-
tures in concentrated suspensions with different viscosities.
In turn, it is also possible to reduce the viscosity of a highly
concentrated suspension by optimizing the size distribution
(i.e., making it more polydisperse).
PELLETIZATION TECHNIQUES
Introduction
The nanonization of drugs results in general in a liquid product
from most of the techniques described in this chapter. These
nanosuspensions have shown excellent long-term stability
without Ostwald ripening or chemical alteration (9,45). In
some special cases, the nanosuspension can be directly used
as a final product, for instance, as pediatric or geriatric dosage
forms if the drug absorption rate is limited only by the solubi-
lity and dissolution rate of the drug. Apart from this—in case of
oral administration—a dry dosage form is clearly preferred for
the reasons of convenience (i.e., marketing aspects). There are
also other cases in which a more sophisticated dosage form is
needed, e.g., to prevent the drug from degradation, to achieve
a controlled drug release or to enable better drug targeting. For
these reasons, the nanosuspension can be transformed into a
solid dosage form by using established techniques, like pelleti-
zation, granulation, spray drying, or lyophilization.
Manufacturing of Nanoparticles 41
Many different pelletization techniques are known, but
the most commonly used techniques are the extrusion–spher-
onization and the drug layering onto sugar spheres. The choice
of the pellet type depends on the required drug content, the
drug properties, and the available technical equipment. Irre-
spective of the pelletization technique applied, a multiparticu-
late dosage form with distinct advantages in comparison to
single-unit dosage forms will be obtained. Multiparticulate
dosage forms, such as coated pellet systems, show a faster
and more predictable gastric emptying and more uniform drug
distribution in the GI tract with less inter- and intraindividual
variability in bioavailability (46). A broad distribution of the
pellets in the gut lumen can enhance the complete redisper-
sion of the nanoparticles from the final solid dosage form.
The incorporation of drug nanocrystals in the various pellet
systems is schematically shown in Figure 11.
Matrix Pellet Preparation
Aqueous nanosuspensions can be mixed with matrix materials
(fillers such as MCC, lactose, or starch); in addition, the
nanosuspension works as a binder and wetting fluid for the
Figure 11 Schematic drawing of different pellet types containing
drug nanocrystals: coated pellet with a sugar bead as core material
and a compact layer consisting of a binder/drug nanocrystal layer
(left) and a coated matrix pellet with a matrix consisting of a
binder/drug nanocrystal mixture as core material (right).
42 Mu€ller et al.
extrusion process (27,47–49). Binders like gelatine, HPMC,
chitosans, or other polymers can be added to the nanosus-
pension before the high-pressure homogenization, which sim-
plifies the production process. Alternatively, they can be
dispersed in the produced nanosuspension after the high-
pressure homogenization. On the one hand these binders are
necessary for the extrusion process, but on the other they
can also positively influence the properties of the nanosuspen-
sion or the nanoparticles. The increased viscosity of the nano-
suspension leads to an increased physical stability of the
nanosuspension with a decreased tendency of sedimentation,
an important factor to obtain reproducible drug content in
the final product. Another important point is the possibility
to change the zeta potential of the drug nanocrystals by using
charged polymers (i.e., chitosan or alginate) to increase the
nanosuspension stability under the GI conditions and to
achieve better drug targeting (50–53). If the nanosuspension
is used as described earlier, one limitation is the maximum
achievable drug content of the final product.
In order to overcome this problem an additional drying
step, such as spray drying, has to be performed to obtain a
fine nanocrystalline powder. This powder can be admixed to
the matrix material to obtain a mass highly loaded with drug
nanocrystals and ready for the extrusion and subsequent
spheronization. Afterward a coating can be applied to the
matrix cores to modify their drug release properties. Figure
12 shows the major steps in the production of matrix pellets
containing drug nanocrystals. A detailed view on these pellets
is given in Figure 13.
Pellet Preparation by Nanosuspension Layering
An alternative way to transfer a prior produced nanosuspen-
sion into a pellet formulation is the suspension layering onto
sugar spheres (54). The binders that are necessary for this
process can also be added before the high-pressure homogeni-
zation process resulting in the improved nanosuspension
properties mentioned earlier. A schematic production process
is shown in Figure 14.
Manufacturing of Nanoparticles 43
The most important difference between the matrix cores
and layered cores is the different drug loading. For the pro-
duction of matrix cores from an aqueous nanosuspension
without an additional drying step, the drug loading is limited
to 4.5%, based on Equation (1), whereas the drug loading of
layered cores can be raised by increasing the layering level
almost without any limitations. (J. Moschwitzer and R. H.
Muller, submitted for publication.)
Drug contentðMCÞ ¼
30% drug contentðNÞÂ15gN
100gðtotal pellet massÞ
¼ 4:5% ð1Þ
Calculated maximal achievable drug content in matrix cores
without additional drying steps: MC ¼ matrix core, N ¼ nano-
suspension (30% is an achievable drug concentration in the
nanosuspension, of course depending on the formulation
and equipment).
Figure 12 Production of drug nanocrystal-loaded matrix cores:
the drug nanosuspension obtained by high-pressure homogeniza-
tion (Micron Lab 40) is admixed to the matrix material, pellets
are prepared by extrusion–spheronization and can be subsequently
coated with polymers with the same equipment to modify the drug
release properties.
44 Mu€ller et al.
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development
Pharmaceutical Textbooks Covering Drug Development

More Related Content

What's hot

Polygonum Persicaria (Linn.) and its Active Principle have a hepatoprotective...
Polygonum Persicaria (Linn.) and its Active Principle have a hepatoprotective...Polygonum Persicaria (Linn.) and its Active Principle have a hepatoprotective...
Polygonum Persicaria (Linn.) and its Active Principle have a hepatoprotective...AI Publications
 
An observational descriptive study of pattern of pathological changes in live...
An observational descriptive study of pattern of pathological changes in live...An observational descriptive study of pattern of pathological changes in live...
An observational descriptive study of pattern of pathological changes in live...AI Publications
 
2.Sagar Goda Biological classification system (BCS); its significance on diss...
2.Sagar Goda Biological classification system (BCS); its significance on diss...2.Sagar Goda Biological classification system (BCS); its significance on diss...
2.Sagar Goda Biological classification system (BCS); its significance on diss...Sagar Goda
 
Bcs classification system
Bcs classification systemBcs classification system
Bcs classification systemharibabu polam
 
Biopharmaceutical Classification System
 Biopharmaceutical Classification System Biopharmaceutical Classification System
Biopharmaceutical Classification SystemVaishnaviBhosale6
 
Biopharmaceutics classification system
Biopharmaceutics classification systemBiopharmaceutics classification system
Biopharmaceutics classification systemReshma Fathima .K
 
Biopharmaceutics Classification System
Biopharmaceutics Classification SystemBiopharmaceutics Classification System
Biopharmaceutics Classification SystemShereen Shehata
 
Drug development
Drug developmentDrug development
Drug developmentGehad Sayed
 
Polyherbal formulation development for anti asthmatic activity
Polyherbal formulation development for anti asthmatic activityPolyherbal formulation development for anti asthmatic activity
Polyherbal formulation development for anti asthmatic activitypharmaindexing
 
Toxicokinetics of pharmdians
Toxicokinetics of pharmdians Toxicokinetics of pharmdians
Toxicokinetics of pharmdians Sugunapharmd
 
Parenteral controlled release drug delivery system - by varsha phirke
Parenteral controlled release drug delivery system - by varsha phirkeParenteral controlled release drug delivery system - by varsha phirke
Parenteral controlled release drug delivery system - by varsha phirkeVarsha Phirke
 
Toxicokinetics or pharmacokinetics
Toxicokinetics or pharmacokineticsToxicokinetics or pharmacokinetics
Toxicokinetics or pharmacokineticsMuhammad Amir Sohail
 
Gastroretentive Drug Delivery system
Gastroretentive Drug Delivery systemGastroretentive Drug Delivery system
Gastroretentive Drug Delivery systemsahebrao Boraste
 
Application of biopharmaceutics classification system in formulation developm...
Application of biopharmaceutics classification system in formulation developm...Application of biopharmaceutics classification system in formulation developm...
Application of biopharmaceutics classification system in formulation developm...Surang Judistprasert
 
Bioequivalence
BioequivalenceBioequivalence
BioequivalenceGaurav Kr
 
Bcs classification system
Bcs classification systemBcs classification system
Bcs classification systemGaurav Kr
 

What's hot (20)

Methods for studying drug uptake
Methods for studying drug uptakeMethods for studying drug uptake
Methods for studying drug uptake
 
Polygonum Persicaria (Linn.) and its Active Principle have a hepatoprotective...
Polygonum Persicaria (Linn.) and its Active Principle have a hepatoprotective...Polygonum Persicaria (Linn.) and its Active Principle have a hepatoprotective...
Polygonum Persicaria (Linn.) and its Active Principle have a hepatoprotective...
 
An observational descriptive study of pattern of pathological changes in live...
An observational descriptive study of pattern of pathological changes in live...An observational descriptive study of pattern of pathological changes in live...
An observational descriptive study of pattern of pathological changes in live...
 
2.Sagar Goda Biological classification system (BCS); its significance on diss...
2.Sagar Goda Biological classification system (BCS); its significance on diss...2.Sagar Goda Biological classification system (BCS); its significance on diss...
2.Sagar Goda Biological classification system (BCS); its significance on diss...
 
Bcs classification system
Bcs classification systemBcs classification system
Bcs classification system
 
Biopharmaceutical Classification System
 Biopharmaceutical Classification System Biopharmaceutical Classification System
Biopharmaceutical Classification System
 
Biopharmaceutics classification system
Biopharmaceutics classification systemBiopharmaceutics classification system
Biopharmaceutics classification system
 
Biopharmaceutics Classification System
Biopharmaceutics Classification SystemBiopharmaceutics Classification System
Biopharmaceutics Classification System
 
Drug development
Drug developmentDrug development
Drug development
 
Polyherbal formulation development for anti asthmatic activity
Polyherbal formulation development for anti asthmatic activityPolyherbal formulation development for anti asthmatic activity
Polyherbal formulation development for anti asthmatic activity
 
Toxicokinetics of pharmdians
Toxicokinetics of pharmdians Toxicokinetics of pharmdians
Toxicokinetics of pharmdians
 
Parenteral controlled release drug delivery system - by varsha phirke
Parenteral controlled release drug delivery system - by varsha phirkeParenteral controlled release drug delivery system - by varsha phirke
Parenteral controlled release drug delivery system - by varsha phirke
 
Toxicokinetics or pharmacokinetics
Toxicokinetics or pharmacokineticsToxicokinetics or pharmacokinetics
Toxicokinetics or pharmacokinetics
 
Gastroretentive Drug Delivery system
Gastroretentive Drug Delivery systemGastroretentive Drug Delivery system
Gastroretentive Drug Delivery system
 
Application of biopharmaceutics classification system in formulation developm...
Application of biopharmaceutics classification system in formulation developm...Application of biopharmaceutics classification system in formulation developm...
Application of biopharmaceutics classification system in formulation developm...
 
1557 full
1557 full1557 full
1557 full
 
Phototoxicity copy
Phototoxicity   copyPhototoxicity   copy
Phototoxicity copy
 
Bioequivalence
BioequivalenceBioequivalence
Bioequivalence
 
Bcs
BcsBcs
Bcs
 
Bcs classification system
Bcs classification systemBcs classification system
Bcs classification system
 

Viewers also liked

Brazil, medicina tropical y parasitología
Brazil, medicina tropical y parasitologíaBrazil, medicina tropical y parasitología
Brazil, medicina tropical y parasitologíaEnzo_camino
 
12 Kesalahan Fatal Penulis Pemula Buku
12 Kesalahan Fatal Penulis Pemula Buku12 Kesalahan Fatal Penulis Pemula Buku
12 Kesalahan Fatal Penulis Pemula BukuSainal Edi Kamal
 
Meditory Journal (Vol. 1, No. 2, Desember 2013)
Meditory Journal (Vol. 1, No. 2, Desember 2013)Meditory Journal (Vol. 1, No. 2, Desember 2013)
Meditory Journal (Vol. 1, No. 2, Desember 2013)Sainal Edi Kamal
 
New Pharmaceutical, Nutraceutical dan Industrial Products
New Pharmaceutical, Nutraceutical dan Industrial ProductsNew Pharmaceutical, Nutraceutical dan Industrial Products
New Pharmaceutical, Nutraceutical dan Industrial ProductsSainal Edi Kamal
 
Clinical parasitology
Clinical parasitologyClinical parasitology
Clinical parasitologybadival
 
Fundamentals of Physiological Psychology by Author Carlson, Neil R.
Fundamentals of Physiological Psychology by Author Carlson, Neil R.Fundamentals of Physiological Psychology by Author Carlson, Neil R.
Fundamentals of Physiological Psychology by Author Carlson, Neil R.NIMHANS
 

Viewers also liked (10)

Brazil, medicina tropical y parasitología
Brazil, medicina tropical y parasitologíaBrazil, medicina tropical y parasitología
Brazil, medicina tropical y parasitología
 
Pain Current Treatment
Pain Current TreatmentPain Current Treatment
Pain Current Treatment
 
12 Kesalahan Fatal Penulis Pemula Buku
12 Kesalahan Fatal Penulis Pemula Buku12 Kesalahan Fatal Penulis Pemula Buku
12 Kesalahan Fatal Penulis Pemula Buku
 
Meditory Journal (Vol. 1, No. 2, Desember 2013)
Meditory Journal (Vol. 1, No. 2, Desember 2013)Meditory Journal (Vol. 1, No. 2, Desember 2013)
Meditory Journal (Vol. 1, No. 2, Desember 2013)
 
New Pharmaceutical, Nutraceutical dan Industrial Products
New Pharmaceutical, Nutraceutical dan Industrial ProductsNew Pharmaceutical, Nutraceutical dan Industrial Products
New Pharmaceutical, Nutraceutical dan Industrial Products
 
Handbook of implant dentistry
Handbook of implant dentistryHandbook of implant dentistry
Handbook of implant dentistry
 
Bahan ajar alat kesehatan
Bahan ajar alat kesehatanBahan ajar alat kesehatan
Bahan ajar alat kesehatan
 
Clinical parasitology
Clinical parasitologyClinical parasitology
Clinical parasitology
 
Fundamentals of Physiological Psychology by Author Carlson, Neil R.
Fundamentals of Physiological Psychology by Author Carlson, Neil R.Fundamentals of Physiological Psychology by Author Carlson, Neil R.
Fundamentals of Physiological Psychology by Author Carlson, Neil R.
 
Plants That Fight Cancer
Plants That Fight CancerPlants That Fight Cancer
Plants That Fight Cancer
 

Similar to Pharmaceutical Textbooks Covering Drug Development

14159328-Good-Design-Practices-For-GMP-Pharmaceutical-Facilities
14159328-Good-Design-Practices-For-GMP-Pharmaceutical-Facilities14159328-Good-Design-Practices-For-GMP-Pharmaceutical-Facilities
14159328-Good-Design-Practices-For-GMP-Pharmaceutical-FacilitiesRaquel Pellicier
 
Good Design Practices for GMP Pharmaceutical Facilities.pdf
Good Design Practices for GMP Pharmaceutical Facilities.pdfGood Design Practices for GMP Pharmaceutical Facilities.pdf
Good Design Practices for GMP Pharmaceutical Facilities.pdfAlok Sharma
 
Pharmaceutical process scale up
Pharmaceutical process scale upPharmaceutical process scale up
Pharmaceutical process scale upJayantPatil
 
Drug-Drug Interactions.pdf
Drug-Drug Interactions.pdfDrug-Drug Interactions.pdf
Drug-Drug Interactions.pdfNorhanKhaled15
 
How to get into ICT MTECH .Entrance Test for M. Tech (Pharma) pptx
How to get into ICT MTECH .Entrance Test for M. Tech (Pharma) pptxHow to get into ICT MTECH .Entrance Test for M. Tech (Pharma) pptx
How to get into ICT MTECH .Entrance Test for M. Tech (Pharma) pptxPayaamvohra1
 
RATIONAL USE OF Medicine2.pptx
RATIONAL USE OF Medicine2.pptxRATIONAL USE OF Medicine2.pptx
RATIONAL USE OF Medicine2.pptxStevenABaqrEgili
 
1. Unit I - new drug discovery and development.
1. Unit I - new drug discovery and development.1. Unit I - new drug discovery and development.
1. Unit I - new drug discovery and development.Audumbar Mali
 
Editorial infectious disease and antimicrobial agents the best way to use a l...
Editorial infectious disease and antimicrobial agents the best way to use a l...Editorial infectious disease and antimicrobial agents the best way to use a l...
Editorial infectious disease and antimicrobial agents the best way to use a l...M. Luisetto Pharm.D.Spec. Pharmacology
 
Gjpps global journal of pharmacy and pharmaceutical sciences luisetto m infe...
Gjpps global journal of pharmacy and  pharmaceutical sciences luisetto m infe...Gjpps global journal of pharmacy and  pharmaceutical sciences luisetto m infe...
Gjpps global journal of pharmacy and pharmaceutical sciences luisetto m infe...M. Luisetto Pharm.D.Spec. Pharmacology
 
Scope of pharmacology
Scope of pharmacology Scope of pharmacology
Scope of pharmacology M R S
 
Clinical Trials.pptx
Clinical Trials.pptxClinical Trials.pptx
Clinical Trials.pptxrehanrustam1
 
Phases_of_Drug_Discovery_(1)[1].pdf
Phases_of_Drug_Discovery_(1)[1].pdfPhases_of_Drug_Discovery_(1)[1].pdf
Phases_of_Drug_Discovery_(1)[1].pdfMrVIP4
 
Gen Pharmacology Intro Bds
Gen Pharmacology Intro BdsGen Pharmacology Intro Bds
Gen Pharmacology Intro BdsRathnakar U P
 

Similar to Pharmaceutical Textbooks Covering Drug Development (20)

14159328-Good-Design-Practices-For-GMP-Pharmaceutical-Facilities
14159328-Good-Design-Practices-For-GMP-Pharmaceutical-Facilities14159328-Good-Design-Practices-For-GMP-Pharmaceutical-Facilities
14159328-Good-Design-Practices-For-GMP-Pharmaceutical-Facilities
 
Good Design Practices for GMP Pharmaceutical Facilities.pdf
Good Design Practices for GMP Pharmaceutical Facilities.pdfGood Design Practices for GMP Pharmaceutical Facilities.pdf
Good Design Practices for GMP Pharmaceutical Facilities.pdf
 
Pharmaceutical process scale up
Pharmaceutical process scale upPharmaceutical process scale up
Pharmaceutical process scale up
 
Drug-Drug Interactions.pdf
Drug-Drug Interactions.pdfDrug-Drug Interactions.pdf
Drug-Drug Interactions.pdf
 
Dk1719 fm
Dk1719 fmDk1719 fm
Dk1719 fm
 
Pharmacy book
Pharmacy bookPharmacy book
Pharmacy book
 
How to get into ICT MTECH .Entrance Test for M. Tech (Pharma) pptx
How to get into ICT MTECH .Entrance Test for M. Tech (Pharma) pptxHow to get into ICT MTECH .Entrance Test for M. Tech (Pharma) pptx
How to get into ICT MTECH .Entrance Test for M. Tech (Pharma) pptx
 
12 m4wpy142
12 m4wpy14212 m4wpy142
12 m4wpy142
 
Pharmacology
PharmacologyPharmacology
Pharmacology
 
Holly Fernandez Lynch, "FDA in the
Holly Fernandez Lynch, "FDA in the Holly Fernandez Lynch, "FDA in the
Holly Fernandez Lynch, "FDA in the
 
RATIONAL USE OF Medicine2.pptx
RATIONAL USE OF Medicine2.pptxRATIONAL USE OF Medicine2.pptx
RATIONAL USE OF Medicine2.pptx
 
Introduction to Pharmacology
Introduction to PharmacologyIntroduction to Pharmacology
Introduction to Pharmacology
 
Clinical Trials - An Introduction
Clinical Trials - An IntroductionClinical Trials - An Introduction
Clinical Trials - An Introduction
 
1. Unit I - new drug discovery and development.
1. Unit I - new drug discovery and development.1. Unit I - new drug discovery and development.
1. Unit I - new drug discovery and development.
 
Editorial infectious disease and antimicrobial agents the best way to use a l...
Editorial infectious disease and antimicrobial agents the best way to use a l...Editorial infectious disease and antimicrobial agents the best way to use a l...
Editorial infectious disease and antimicrobial agents the best way to use a l...
 
Gjpps global journal of pharmacy and pharmaceutical sciences luisetto m infe...
Gjpps global journal of pharmacy and  pharmaceutical sciences luisetto m infe...Gjpps global journal of pharmacy and  pharmaceutical sciences luisetto m infe...
Gjpps global journal of pharmacy and pharmaceutical sciences luisetto m infe...
 
Scope of pharmacology
Scope of pharmacology Scope of pharmacology
Scope of pharmacology
 
Clinical Trials.pptx
Clinical Trials.pptxClinical Trials.pptx
Clinical Trials.pptx
 
Phases_of_Drug_Discovery_(1)[1].pdf
Phases_of_Drug_Discovery_(1)[1].pdfPhases_of_Drug_Discovery_(1)[1].pdf
Phases_of_Drug_Discovery_(1)[1].pdf
 
Gen Pharmacology Intro Bds
Gen Pharmacology Intro BdsGen Pharmacology Intro Bds
Gen Pharmacology Intro Bds
 

More from Sainal Edi Kamal

Badan Pengawas Obat dan Makanan - Republik Indonesia.pdf
Badan Pengawas Obat dan Makanan - Republik Indonesia.pdfBadan Pengawas Obat dan Makanan - Republik Indonesia.pdf
Badan Pengawas Obat dan Makanan - Republik Indonesia.pdfSainal Edi Kamal
 
Pedoman-Operasional-BKD-2021-Kirim.pdf
Pedoman-Operasional-BKD-2021-Kirim.pdfPedoman-Operasional-BKD-2021-Kirim.pdf
Pedoman-Operasional-BKD-2021-Kirim.pdfSainal Edi Kamal
 
Rekomendasi IDAI Gagal Ginjal Akut pada Anak yang blm diketahui penyebabnya_1...
Rekomendasi IDAI Gagal Ginjal Akut pada Anak yang blm diketahui penyebabnya_1...Rekomendasi IDAI Gagal Ginjal Akut pada Anak yang blm diketahui penyebabnya_1...
Rekomendasi IDAI Gagal Ginjal Akut pada Anak yang blm diketahui penyebabnya_1...Sainal Edi Kamal
 
Standar Laboratorium Farmasi
Standar Laboratorium FarmasiStandar Laboratorium Farmasi
Standar Laboratorium FarmasiSainal Edi Kamal
 
Green Tea. Health Benefits and Applications
Green Tea. Health Benefits and ApplicationsGreen Tea. Health Benefits and Applications
Green Tea. Health Benefits and ApplicationsSainal Edi Kamal
 
Permenkes No. 84 Tahun 2014 Tentang DAK 2015
Permenkes No. 84 Tahun 2014 Tentang DAK 2015Permenkes No. 84 Tahun 2014 Tentang DAK 2015
Permenkes No. 84 Tahun 2014 Tentang DAK 2015Sainal Edi Kamal
 
Permenkes No. 34 Tahun 2014 Tentang PBF
Permenkes No. 34 Tahun 2014 Tentang PBFPermenkes No. 34 Tahun 2014 Tentang PBF
Permenkes No. 34 Tahun 2014 Tentang PBFSainal Edi Kamal
 
Permenkes No. 161 Tahun 2010 Tentang Registrasi Tenaga Kesehatan
Permenkes No. 161 Tahun 2010 Tentang Registrasi Tenaga KesehatanPermenkes No. 161 Tahun 2010 Tentang Registrasi Tenaga Kesehatan
Permenkes No. 161 Tahun 2010 Tentang Registrasi Tenaga KesehatanSainal Edi Kamal
 
Permenkes No.1799 Tahun 2010 Tentang Industri Farmasi
Permenkes No.1799 Tahun 2010 Tentang Industri FarmasiPermenkes No.1799 Tahun 2010 Tentang Industri Farmasi
Permenkes No.1799 Tahun 2010 Tentang Industri FarmasiSainal Edi Kamal
 
Permenkes No.028 Tahun 2011 Tentang Klinik
Permenkes No.028 Tahun 2011 Tentang KlinikPermenkes No.028 Tahun 2011 Tentang Klinik
Permenkes No.028 Tahun 2011 Tentang KlinikSainal Edi Kamal
 
Permenkes No. 1148 Tahun 2011 Tentang PBF
Permenkes No. 1148 Tahun 2011 Tentang PBFPermenkes No. 1148 Tahun 2011 Tentang PBF
Permenkes No. 1148 Tahun 2011 Tentang PBFSainal Edi Kamal
 
Permenkes No. 889 Tahun 2011 Tentang Registrasi, Izin Praktek dan Izin Kerja ...
Permenkes No. 889 Tahun 2011 Tentang Registrasi, Izin Praktek dan Izin Kerja ...Permenkes No. 889 Tahun 2011 Tentang Registrasi, Izin Praktek dan Izin Kerja ...
Permenkes No. 889 Tahun 2011 Tentang Registrasi, Izin Praktek dan Izin Kerja ...Sainal Edi Kamal
 
Permenkes No. 006 ttg industri dan usaha obat tradisional
Permenkes No. 006 ttg industri dan usaha obat tradisionalPermenkes No. 006 ttg industri dan usaha obat tradisional
Permenkes No. 006 ttg industri dan usaha obat tradisionalSainal Edi Kamal
 
Cerdas Memilih Obat dan Memilih Penyakit
Cerdas Memilih Obat dan Memilih PenyakitCerdas Memilih Obat dan Memilih Penyakit
Cerdas Memilih Obat dan Memilih PenyakitSainal Edi Kamal
 
Jadi Dokter untuk Diri Sendiri
Jadi Dokter untuk Diri SendiriJadi Dokter untuk Diri Sendiri
Jadi Dokter untuk Diri SendiriSainal Edi Kamal
 
Bijak Memanfaatkan Antibiotik
Bijak Memanfaatkan AntibiotikBijak Memanfaatkan Antibiotik
Bijak Memanfaatkan AntibiotikSainal Edi Kamal
 

More from Sainal Edi Kamal (20)

Badan Pengawas Obat dan Makanan - Republik Indonesia.pdf
Badan Pengawas Obat dan Makanan - Republik Indonesia.pdfBadan Pengawas Obat dan Makanan - Republik Indonesia.pdf
Badan Pengawas Obat dan Makanan - Republik Indonesia.pdf
 
Pedoman-Operasional-BKD-2021-Kirim.pdf
Pedoman-Operasional-BKD-2021-Kirim.pdfPedoman-Operasional-BKD-2021-Kirim.pdf
Pedoman-Operasional-BKD-2021-Kirim.pdf
 
Rekomendasi IDAI Gagal Ginjal Akut pada Anak yang blm diketahui penyebabnya_1...
Rekomendasi IDAI Gagal Ginjal Akut pada Anak yang blm diketahui penyebabnya_1...Rekomendasi IDAI Gagal Ginjal Akut pada Anak yang blm diketahui penyebabnya_1...
Rekomendasi IDAI Gagal Ginjal Akut pada Anak yang blm diketahui penyebabnya_1...
 
Farmakologi Dasar
Farmakologi DasarFarmakologi Dasar
Farmakologi Dasar
 
Bahan Ajar Alat Kesehatan
Bahan Ajar Alat KesehatanBahan Ajar Alat Kesehatan
Bahan Ajar Alat Kesehatan
 
Standar Laboratorium Farmasi
Standar Laboratorium FarmasiStandar Laboratorium Farmasi
Standar Laboratorium Farmasi
 
Farmakologi Dasar
Farmakologi DasarFarmakologi Dasar
Farmakologi Dasar
 
Green Tea. Health Benefits and Applications
Green Tea. Health Benefits and ApplicationsGreen Tea. Health Benefits and Applications
Green Tea. Health Benefits and Applications
 
Permenkes No. 84 Tahun 2014 Tentang DAK 2015
Permenkes No. 84 Tahun 2014 Tentang DAK 2015Permenkes No. 84 Tahun 2014 Tentang DAK 2015
Permenkes No. 84 Tahun 2014 Tentang DAK 2015
 
Permenkes No. 34 Tahun 2014 Tentang PBF
Permenkes No. 34 Tahun 2014 Tentang PBFPermenkes No. 34 Tahun 2014 Tentang PBF
Permenkes No. 34 Tahun 2014 Tentang PBF
 
Permenkes No. 161 Tahun 2010 Tentang Registrasi Tenaga Kesehatan
Permenkes No. 161 Tahun 2010 Tentang Registrasi Tenaga KesehatanPermenkes No. 161 Tahun 2010 Tentang Registrasi Tenaga Kesehatan
Permenkes No. 161 Tahun 2010 Tentang Registrasi Tenaga Kesehatan
 
Permenkes No.1799 Tahun 2010 Tentang Industri Farmasi
Permenkes No.1799 Tahun 2010 Tentang Industri FarmasiPermenkes No.1799 Tahun 2010 Tentang Industri Farmasi
Permenkes No.1799 Tahun 2010 Tentang Industri Farmasi
 
Permenkes No.028 Tahun 2011 Tentang Klinik
Permenkes No.028 Tahun 2011 Tentang KlinikPermenkes No.028 Tahun 2011 Tentang Klinik
Permenkes No.028 Tahun 2011 Tentang Klinik
 
Permenkes No. 1148 Tahun 2011 Tentang PBF
Permenkes No. 1148 Tahun 2011 Tentang PBFPermenkes No. 1148 Tahun 2011 Tentang PBF
Permenkes No. 1148 Tahun 2011 Tentang PBF
 
Permenkes No. 889 Tahun 2011 Tentang Registrasi, Izin Praktek dan Izin Kerja ...
Permenkes No. 889 Tahun 2011 Tentang Registrasi, Izin Praktek dan Izin Kerja ...Permenkes No. 889 Tahun 2011 Tentang Registrasi, Izin Praktek dan Izin Kerja ...
Permenkes No. 889 Tahun 2011 Tentang Registrasi, Izin Praktek dan Izin Kerja ...
 
Permenkes No. 006 ttg industri dan usaha obat tradisional
Permenkes No. 006 ttg industri dan usaha obat tradisionalPermenkes No. 006 ttg industri dan usaha obat tradisional
Permenkes No. 006 ttg industri dan usaha obat tradisional
 
Cerdas Memilih Obat dan Memilih Penyakit
Cerdas Memilih Obat dan Memilih PenyakitCerdas Memilih Obat dan Memilih Penyakit
Cerdas Memilih Obat dan Memilih Penyakit
 
Jadi Dokter untuk Diri Sendiri
Jadi Dokter untuk Diri SendiriJadi Dokter untuk Diri Sendiri
Jadi Dokter untuk Diri Sendiri
 
Bijak Memanfaatkan Antibiotik
Bijak Memanfaatkan AntibiotikBijak Memanfaatkan Antibiotik
Bijak Memanfaatkan Antibiotik
 
Farmakologi Kebidanan
Farmakologi KebidananFarmakologi Kebidanan
Farmakologi Kebidanan
 

Recently uploaded

Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...Krashi Coaching
 
Measures of Dispersion and Variability: Range, QD, AD and SD
Measures of Dispersion and Variability: Range, QD, AD and SDMeasures of Dispersion and Variability: Range, QD, AD and SD
Measures of Dispersion and Variability: Range, QD, AD and SDThiyagu K
 
JAPAN: ORGANISATION OF PMDA, PHARMACEUTICAL LAWS & REGULATIONS, TYPES OF REGI...
JAPAN: ORGANISATION OF PMDA, PHARMACEUTICAL LAWS & REGULATIONS, TYPES OF REGI...JAPAN: ORGANISATION OF PMDA, PHARMACEUTICAL LAWS & REGULATIONS, TYPES OF REGI...
JAPAN: ORGANISATION OF PMDA, PHARMACEUTICAL LAWS & REGULATIONS, TYPES OF REGI...anjaliyadav012327
 
Measures of Central Tendency: Mean, Median and Mode
Measures of Central Tendency: Mean, Median and ModeMeasures of Central Tendency: Mean, Median and Mode
Measures of Central Tendency: Mean, Median and ModeThiyagu K
 
Accessible design: Minimum effort, maximum impact
Accessible design: Minimum effort, maximum impactAccessible design: Minimum effort, maximum impact
Accessible design: Minimum effort, maximum impactdawncurless
 
1029 - Danh muc Sach Giao Khoa 10 . pdf
1029 -  Danh muc Sach Giao Khoa 10 . pdf1029 -  Danh muc Sach Giao Khoa 10 . pdf
1029 - Danh muc Sach Giao Khoa 10 . pdfQucHHunhnh
 
Russian Call Girls in Andheri Airport Mumbai WhatsApp 9167673311 💞 Full Nigh...
Russian Call Girls in Andheri Airport Mumbai WhatsApp  9167673311 💞 Full Nigh...Russian Call Girls in Andheri Airport Mumbai WhatsApp  9167673311 💞 Full Nigh...
Russian Call Girls in Andheri Airport Mumbai WhatsApp 9167673311 💞 Full Nigh...Pooja Nehwal
 
Call Girls in Dwarka Mor Delhi Contact Us 9654467111
Call Girls in Dwarka Mor Delhi Contact Us 9654467111Call Girls in Dwarka Mor Delhi Contact Us 9654467111
Call Girls in Dwarka Mor Delhi Contact Us 9654467111Sapana Sha
 
Beyond the EU: DORA and NIS 2 Directive's Global Impact
Beyond the EU: DORA and NIS 2 Directive's Global ImpactBeyond the EU: DORA and NIS 2 Directive's Global Impact
Beyond the EU: DORA and NIS 2 Directive's Global ImpactPECB
 
A Critique of the Proposed National Education Policy Reform
A Critique of the Proposed National Education Policy ReformA Critique of the Proposed National Education Policy Reform
A Critique of the Proposed National Education Policy ReformChameera Dedduwage
 
Paris 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityParis 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityGeoBlogs
 
APM Welcome, APM North West Network Conference, Synergies Across Sectors
APM Welcome, APM North West Network Conference, Synergies Across SectorsAPM Welcome, APM North West Network Conference, Synergies Across Sectors
APM Welcome, APM North West Network Conference, Synergies Across SectorsAssociation for Project Management
 
Mastering the Unannounced Regulatory Inspection
Mastering the Unannounced Regulatory InspectionMastering the Unannounced Regulatory Inspection
Mastering the Unannounced Regulatory InspectionSafetyChain Software
 
Grant Readiness 101 TechSoup and Remy Consulting
Grant Readiness 101 TechSoup and Remy ConsultingGrant Readiness 101 TechSoup and Remy Consulting
Grant Readiness 101 TechSoup and Remy ConsultingTechSoup
 
The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13Steve Thomason
 
1029-Danh muc Sach Giao Khoa khoi 6.pdf
1029-Danh muc Sach Giao Khoa khoi  6.pdf1029-Danh muc Sach Giao Khoa khoi  6.pdf
1029-Danh muc Sach Giao Khoa khoi 6.pdfQucHHunhnh
 
mini mental status format.docx
mini    mental       status     format.docxmini    mental       status     format.docx
mini mental status format.docxPoojaSen20
 

Recently uploaded (20)

Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
 
Measures of Dispersion and Variability: Range, QD, AD and SD
Measures of Dispersion and Variability: Range, QD, AD and SDMeasures of Dispersion and Variability: Range, QD, AD and SD
Measures of Dispersion and Variability: Range, QD, AD and SD
 
JAPAN: ORGANISATION OF PMDA, PHARMACEUTICAL LAWS & REGULATIONS, TYPES OF REGI...
JAPAN: ORGANISATION OF PMDA, PHARMACEUTICAL LAWS & REGULATIONS, TYPES OF REGI...JAPAN: ORGANISATION OF PMDA, PHARMACEUTICAL LAWS & REGULATIONS, TYPES OF REGI...
JAPAN: ORGANISATION OF PMDA, PHARMACEUTICAL LAWS & REGULATIONS, TYPES OF REGI...
 
Measures of Central Tendency: Mean, Median and Mode
Measures of Central Tendency: Mean, Median and ModeMeasures of Central Tendency: Mean, Median and Mode
Measures of Central Tendency: Mean, Median and Mode
 
Accessible design: Minimum effort, maximum impact
Accessible design: Minimum effort, maximum impactAccessible design: Minimum effort, maximum impact
Accessible design: Minimum effort, maximum impact
 
1029 - Danh muc Sach Giao Khoa 10 . pdf
1029 -  Danh muc Sach Giao Khoa 10 . pdf1029 -  Danh muc Sach Giao Khoa 10 . pdf
1029 - Danh muc Sach Giao Khoa 10 . pdf
 
Russian Call Girls in Andheri Airport Mumbai WhatsApp 9167673311 💞 Full Nigh...
Russian Call Girls in Andheri Airport Mumbai WhatsApp  9167673311 💞 Full Nigh...Russian Call Girls in Andheri Airport Mumbai WhatsApp  9167673311 💞 Full Nigh...
Russian Call Girls in Andheri Airport Mumbai WhatsApp 9167673311 💞 Full Nigh...
 
Call Girls in Dwarka Mor Delhi Contact Us 9654467111
Call Girls in Dwarka Mor Delhi Contact Us 9654467111Call Girls in Dwarka Mor Delhi Contact Us 9654467111
Call Girls in Dwarka Mor Delhi Contact Us 9654467111
 
Código Creativo y Arte de Software | Unidad 1
Código Creativo y Arte de Software | Unidad 1Código Creativo y Arte de Software | Unidad 1
Código Creativo y Arte de Software | Unidad 1
 
Beyond the EU: DORA and NIS 2 Directive's Global Impact
Beyond the EU: DORA and NIS 2 Directive's Global ImpactBeyond the EU: DORA and NIS 2 Directive's Global Impact
Beyond the EU: DORA and NIS 2 Directive's Global Impact
 
A Critique of the Proposed National Education Policy Reform
A Critique of the Proposed National Education Policy ReformA Critique of the Proposed National Education Policy Reform
A Critique of the Proposed National Education Policy Reform
 
Mattingly "AI & Prompt Design: Structured Data, Assistants, & RAG"
Mattingly "AI & Prompt Design: Structured Data, Assistants, & RAG"Mattingly "AI & Prompt Design: Structured Data, Assistants, & RAG"
Mattingly "AI & Prompt Design: Structured Data, Assistants, & RAG"
 
Paris 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityParis 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activity
 
APM Welcome, APM North West Network Conference, Synergies Across Sectors
APM Welcome, APM North West Network Conference, Synergies Across SectorsAPM Welcome, APM North West Network Conference, Synergies Across Sectors
APM Welcome, APM North West Network Conference, Synergies Across Sectors
 
Mattingly "AI & Prompt Design: The Basics of Prompt Design"
Mattingly "AI & Prompt Design: The Basics of Prompt Design"Mattingly "AI & Prompt Design: The Basics of Prompt Design"
Mattingly "AI & Prompt Design: The Basics of Prompt Design"
 
Mastering the Unannounced Regulatory Inspection
Mastering the Unannounced Regulatory InspectionMastering the Unannounced Regulatory Inspection
Mastering the Unannounced Regulatory Inspection
 
Grant Readiness 101 TechSoup and Remy Consulting
Grant Readiness 101 TechSoup and Remy ConsultingGrant Readiness 101 TechSoup and Remy Consulting
Grant Readiness 101 TechSoup and Remy Consulting
 
The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13
 
1029-Danh muc Sach Giao Khoa khoi 6.pdf
1029-Danh muc Sach Giao Khoa khoi  6.pdf1029-Danh muc Sach Giao Khoa khoi  6.pdf
1029-Danh muc Sach Giao Khoa khoi 6.pdf
 
mini mental status format.docx
mini    mental       status     format.docxmini    mental       status     format.docx
mini mental status format.docx
 

Pharmaceutical Textbooks Covering Drug Development

  • 1.
  • 2. DRUGS AND THE PHARMACEUTICAL SCIENCES A Series of Textbooks and Monographs 1. Pharmacokinetics, Milo Gibaldi and Donald Perrier 2. Good Manufacturing Practices for Pharmaceuticals: A Plan for Total Quality Control, Sidney H. Willig, Murray M. Tuckerman, and William S. Hitchings IV 3. Microencapsulation, edited by J. R. Nixon 4. Drug Metabolism: Chemical and Biochemical Aspects, Bernard Testa and Peter Jenner 5. New Drugs: Discovery and Development, edited by Alan A. Rubin 6. Sustained and Controlled Release Drug Delivery Systems, edited by Joseph R. Robinson 7. Modern Pharmaceutics, edited by Gilbert S. Banker and Christopher T. Rhodes 8. Prescription Drugs in Short Supply: Case Histories, Michael A. Schwartz 9. Activated Charcoal: Antidotal and Other Medical Uses, David O. Cooney 10. Concepts in Drug Metabolism (in two parts), edited by Peter Jenner and Bernard Testa 11. Pharmaceutical Analysis: Modern Methods (in two parts), edited by James W. Munson 12. Techniques of Solubilization of Drugs, edited by Samuel H. Yalkowsky 13. Orphan Drugs, edited by Fred E. Karch 14. Novel Drug Delivery Systems: Fundamentals, Developmental Concepts, Biomedical Assessments, Yie W. Chien 15. Pharmacokinetics: Second Edition, Revised and Expanded, Milo Gibaldi and Donald Perrier 16. Good Manufacturing Practices for Pharmaceuticals: A Plan for Total Quality Control, Second Edition, Revised and Expanded, Sidney H. Willig, Murray M. Tuckerman, and William S. Hitchings IV 17. Formulation of Veterinary Dosage Forms, edited by Jack Blodinger 18. Dermatological Formulations: Percutaneous Absorption, Brian W. Barry 19. The Clinical Research Process in the Pharmaceutical Industry, edited by Gary M. Matoren 20. Microencapsulation and Related Drug Processes, Patrick B. Deasy 21. Drugs and Nutrients: The Interactive Effects, edited by Daphne A. Roe and T. Colin Campbell 22. Biotechnology of Industrial Antibiotics, Erick J. Vandamme 23. Pharmaceutical Process Validation, edited by Bernard T. Loftus and Robert A. Nash
  • 3. 24. Anticancer and Interferon Agents: Synthesis and Properties, edited by Raphael M. Ottenbrite and George B. Butler 25. Pharmaceutical Statistics: Practical and Clinical Applications, Sanford Bolton 26. Drug Dynamics for Analytical, Clinical, and Biological Chemists, Benjamin J. Gudzinowicz, Burrows T. Younkin, Jr., and Michael J. Gudzinowicz 27. Modern Analysis of Antibiotics, edited by Adjoran Aszalos 28. Solubility and Related Properties, Kenneth C. James 29. Controlled Drug Delivery: Fundamentals and Applications, Second Edition, Revised and Expanded, edited by Joseph R. Robinson and Vincent H. Lee 30. New Drug Approval Process: Clinical and Regulatory Management, edited by Richard A. Guarino 31. Transdermal Controlled Systemic Medications, edited by Yie W. Chien 32. Drug Delivery Devices: Fundamentals and Applications, edited by Praveen Tyle 33. Pharmacokinetics: Regulatory • Industrial • Academic Perspectives, edited by Peter G. Welling and Francis L. S. Tse 34. Clinical Drug Trials and Tribulations, edited by Allen E. Cato 35. Transdermal Drug Delivery: Developmental Issues and Research Initiatives, edited by Jonathan Hadgraft and Richard H. Guy 36. Aqueous Polymeric Coatings for Pharmaceutical Dosage Forms, edited by James W. McGinity 37. Pharmaceutical Pelletization Technology, edited by Isaac Ghebre-Sellassie 38. Good Laboratory Practice Regulations, edited by Allen F. Hirsch 39. Nasal Systemic Drug Delivery, Yie W. Chien, Kenneth S. E. Su, and Shyi-Feu Chang 40. Modern Pharmaceutics: Second Edition, Revised and Expanded, edited by Gilbert S. Banker and Christopher T. Rhodes 41. Specialized Drug Delivery Systems: Manufacturing and Production Technology, edited by Praveen Tyle 42. Topical Drug Delivery Formulations, edited by David W. Osborne and Anton H. Amann 43. Drug Stability: Principles and Practices, Jens T. Carstensen 44. Pharmaceutical Statistics: Practical and Clinical Applications, Second Edition, Revised and Expanded, Sanford Bolton 45. Biodegradable Polymers as Drug Delivery Systems, edited by Mark Chasin and Robert Langer 46. Preclinical Drug Disposition: A Laboratory Handbook, Francis L. S. Tse and James J. Jaffe 47. HPLC in the Pharmaceutical Industry, edited by Godwin W. Fong and Stanley K. Lam
  • 4. 48. Pharmaceutical Bioequivalence, edited by Peter G. Welling, Francis L. S. Tse, and Shrikant V. Dinghe 49. Pharmaceutical Dissolution Testing, Umesh V. Banakar 50. Novel Drug Delivery Systems: Second Edition, Revised and Expanded, Yie W. Chien 51. Managing the Clinical Drug Development Process, David M. Cocchetto and Ronald V. Nardi 52. Good Manufacturing Practices for Pharmaceuticals: A Plan for Total Quality Control, Third Edition, edited by Sidney H. Willig and James R. Stoker 53. Prodrugs: Topical and Ocular Drug Delivery, edited by Kenneth B. Sloan 54. Pharmaceutical Inhalation Aerosol Technology, edited by Anthony J. Hickey 55. Radiopharmaceuticals: Chemistry and Pharmacology, edited by Adrian D. Nunn 56. New Drug Approval Process: Second Edition, Revised and Expanded, edited by Richard A. Guarino 57. Pharmaceutical Process Validation: Second Edition, Revised and Expanded, edited by Ira R. Berry and Robert A. Nash 58. Ophthalmic Drug Delivery Systems, edited by Ashim K. Mitra 59. Pharmaceutical Skin Penetration Enhancement, edited by Kenneth A. Walters and Jonathan Hadgraft 60. Colonic Drug Absorption and Metabolism, edited by Peter R. Bieck 61. Pharmaceutical Particulate Carriers: Therapeutic Applications, edited by Alain Rolland 62. Drug Permeation Enhancement: Theory and Applications, edited by Dean S. Hsieh 63. Glycopeptide Antibiotics, edited by Ramakrishnan Nagarajan 64. Achieving Sterility in Medical and Pharmaceutical Products, Nigel A. Halls 65. Multiparticulate Oral Drug Delivery, edited by Isaac Ghebre-Sellassie 66. Colloidal Drug Delivery Systems, edited by Jörg Kreuter 67. Pharmacokinetics: Regulatory • Industrial • Academic Perspectives, Second Edition, edited by Peter G. Welling and Francis L. S. Tse 68. Drug Stability: Principles and Practices, Second Edition, Revised and Expanded, Jens T. Carstensen 69. Good Laboratory Practice Regulations: Second Edition, Revised and Expanded, edited by Sandy Weinberg 70. Physical Characterization of Pharmaceutical Solids, edited by Harry G. Brittain 71. Pharmaceutical Powder Compaction Technology, edited by Göran Alderborn and Christer Nyström
  • 5. 72. Modern Pharmaceutics: Third Edition, Revised and Expanded, edited by Gilbert S. Banker and Christopher T. Rhodes 73. Microencapsulation: Methods and Industrial Applications, edited by Simon Benita 74. Oral Mucosal Drug Delivery, edited by Michael J. Rathbone 75. Clinical Research in Pharmaceutical Development, edited by Barry Bleidt and Michael Montagne 76. The Drug Development Process: Increasing Efficiency and Cost Effectiveness, edited by Peter G. Welling, Louis Lasagna, and Umesh V. Banakar 77. Microparticulate Systems for the Delivery of Proteins and Vaccines, edited by Smadar Cohen and Howard Bernstein 78. Good Manufacturing Practices for Pharmaceuticals: A Plan for Total Quality Control, Fourth Edition, Revised and Expanded, Sidney H. Willig and James R. Stoker 79. Aqueous Polymeric Coatings for Pharmaceutical Dosage Forms: Second Edition, Revised and Expanded, edited by James W. McGinity 80. Pharmaceutical Statistics: Practical and Clinical Applications, Third Edition, Sanford Bolton 81. Handbook of Pharmaceutical Granulation Technology, edited by Dilip M. Parikh 82. Biotechnology of Antibiotics: Second Edition, Revised and Expanded, edited by William R. Strohl 83. Mechanisms of Transdermal Drug Delivery, edited by Russell O. Potts and Richard H. Guy 84. Pharmaceutical Enzymes, edited by Albert Lauwers and Simon Scharpé 85. Development of Biopharmaceutical Parenteral Dosage Forms, edited by John A. Bontempo 86. Pharmaceutical Project Management, edited by Tony Kennedy 87. Drug Products for Clinical Trials: An International Guide to Formulation • Production • Quality Control, edited by Donald C. Monkhouse and Christopher T. Rhodes 88. Development and Formulation of Veterinary Dosage Forms: Second Edition, Revised and Expanded, edited by Gregory E. Hardee and J. Desmond Baggot 89. Receptor-Based Drug Design, edited by Paul Leff 90. Automation and Validation of Information in Pharmaceutical Processing, edited by Joseph F. deSpautz 91. Dermal Absorption and Toxicity Assessment, edited by Michael S. Roberts and Kenneth A. Walters 92. Pharmaceutical Experimental Design, Gareth A. Lewis, Didier Mathieu, and Roger Phan-Tan-Luu 93. Preparing for FDA Pre-Approval Inspections, edited by Martin D. Hynes III
  • 6. 94. Pharmaceutical Excipients: Characterization by IR, Raman, and NMR Spectroscopy, David E. Bugay and W. Paul Findlay 95. Polymorphism in Pharmaceutical Solids, edited by Harry G. Brittain 96. Freeze-Drying/Lyophilization of Pharmaceutical and Biological Products, edited by Louis Rey and Joan C. May 97. Percutaneous Absorption: Drugs–Cosmetics–Mechanisms–Methodology, Third Edition, Revised and Expanded, edited by Robert L. Bronaugh and Howard I. Maibach 98. Bioadhesive Drug Delivery Systems: Fundamentals, Novel Approaches, and Development, edited by Edith Mathiowitz, Donald E. Chickering III, and Claus-Michael Lehr 99. Protein Formulation and Delivery, edited by Eugene J. McNally 100. New Drug Approval Process: Third Edition, The Global Challenge, edited by Richard A. Guarino 101. Peptide and Protein Drug Analysis, edited by Ronald E. Reid 102. Transport Processes in Pharmaceutical Systems, edited by Gordon L. Amidon, Ping I. Lee, and Elizabeth M. Topp 103. Excipient Toxicity and Safety, edited by Myra L. Weiner and Lois A. Kotkoskie 104. The Clinical Audit in Pharmaceutical Development, edited by Michael R. Hamrell 105. Pharmaceutical Emulsions and Suspensions, edited by Francoise Nielloud and Gilberte Marti-Mestres 106. Oral Drug Absorption: Prediction and Assessment, edited by Jennifer B. Dressman and Hans Lennernäs 107. Drug Stability: Principles and Practices, Third Edition, Revised and Expanded, edited by Jens T. Carstensen and C. T. Rhodes 108. Containment in the Pharmaceutical Industry, edited by James P. Wood 109. Good Manufacturing Practices for Pharmaceuticals: A Plan for Total Quality Control from Manufacturer to Consumer, Fifth Edition, Revised and Expanded, Sidney H. Willig 110. Advanced Pharmaceutical Solids, Jens T. Carstensen 111. Endotoxins: Pyrogens, LAL Testing, and Depyrogenation, Second Edition, Revised and Expanded, Kevin L. Williams 112. Pharmaceutical Process Engineering, Anthony J. Hickey and David Ganderton 113. Pharmacogenomics, edited by Werner Kalow, Urs A. Meyer and Rachel F. Tyndale 114. Handbook of Drug Screening, edited by Ramakrishna Seethala and Prabhavathi B. Fernandes 115. Drug Targeting Technology: Physical • Chemical • Biological Methods, edited by Hans Schreier 116. Drug–Drug Interactions, edited by A. David Rodrigues
  • 7. 117. Handbook of Pharmaceutical Analysis, edited by Lena Ohannesian and Anthony J. Streeter 118. Pharmaceutical Process Scale-Up, edited by Michael Levin 119. Dermatological and Transdermal Formulations, edited by Kenneth A. Walters 120. Clinical Drug Trials and Tribulations: Second Edition, Revised and Expanded, edited by Allen Cato, Lynda Sutton, and Allen Cato III 121. Modern Pharmaceutics: Fourth Edition, Revised and Expanded, edited by Gilbert S. Banker and Christopher T. Rhodes 122. Surfactants and Polymers in Drug Delivery, Martin Malmsten 123. Transdermal Drug Delivery: Second Edition, Revised and Expanded, edited by Richard H. Guy and Jonathan Hadgraft 124. Good Laboratory Practice Regulations: Second Edition, Revised and Expanded, edited by Sandy Weinberg 125. Parenteral Quality Control: Sterility, Pyrogen, Particulate, and Package Integrity Testing: Third Edition, Revised and Expanded, Michael J. Akers, Daniel S. Larrimore, and Dana Morton Guazzo 126. Modified-Release Drug Delivery Technology, edited by Michael J. Rathbone, Jonathan Hadgraft, and Michael S. Roberts 127. Simulation for Designing Clinical Trials: A Pharmacokinetic- Pharmacodynamic Modeling Perspective, edited by Hui C. Kimko and Stephen B. Duffull 128. Affinity Capillary Electrophoresis in Pharmaceutics and Biopharmaceutics, edited by Reinhard H. H. Neubert and Hans-Hermann Rüttinger 129. Pharmaceutical Process Validation: An International Third Edition, Revised and Expanded, edited by Robert A. Nash and Alfred H. Wachter 130. Ophthalmic Drug Delivery Systems: Second Edition, Revised and Expanded, edited by Ashim K. Mitra 131. Pharmaceutical Gene Delivery Systems, edited by Alain Rolland and Sean M. Sullivan 132. Biomarkers in Clinical Drug Development, edited by John C. Bloom and Robert A. Dean 133. Pharmaceutical Extrusion Technology, edited by Isaac Ghebre-Sellassie and Charles Martin 134. Pharmaceutical Inhalation Aerosol Technology: Second Edition, Revised and Expanded, edited by Anthony J. Hickey 135. Pharmaceutical Statistics: Practical and Clinical Applications, Fourth Edition, Sanford Bolton and Charles Bon 136. Compliance Handbook for Pharmaceuticals, Medical Devices, and Biologics, edited by Carmen Medina 137. Freeze-Drying/Lyophilization of Pharmaceutical and Biological Products: Second Edition, Revised and Expanded, edited by Louis Rey and Joan C. May
  • 8. 138. Supercritical Fluid Technology for Drug Product Development, edited by Peter York, Uday B. Kompella, and Boris Y. Shekunov 139. New Drug Approval Process: Fourth Edition, Accelerating Global Registrations, edited by Richard A. Guarino 140. Microbial Contamination Control in Parenteral Manufacturing, edited by Kevin L. Williams 141. New Drug Development: Regulatory Paradigms for Clinical Pharmacology and Biopharmaceutics, edited by Chandrahas G. Sahajwalla 142. Microbial Contamination Control in the Pharmaceutical Industry, edited by Luis Jimenez 143. Generic Drug Product Development: Solid Oral Dosage Forms, edited by Leon Shargel and Izzy Kanfer 144. Introduction to the Pharmaceutical Regulatory Process, edited by Ira R. Berry 145. Drug Delivery to the Oral Cavity: Molecules to Market, edited by Tapash K. Ghosh and William R. Pfister 146. Good Design Practices for GMP Pharmaceutical Facilities, edited by Andrew Signore and Terry Jacobs 147. Drug Products for Clinical Trials, Second Edition, edited by Donald Monkhouse, Charles Carney, and Jim Clark 148. Polymeric Drug Delivery Systems, edited by Glen S. Kwon 149. Injectable Dispersed Systems: Formulation, Processing, and Performance, edited by Diane J. Burgess 150. Laboratory Auditing for Quality and Regulatory Compliance, Donald Singer, Raluca-Ioana Stefan, and Jacobus van Staden 151. Active Pharmaceutical Ingredients: Development, Manufacturing, and Regulation, edited by Stanley Nusim 152. Preclinical Drug Development, edited by Mark C. Rogge and David R. Taft 153. Pharmaceutical Stress Testing: Predicting Drug Degradation, edited by Steven W. Baertschi 154. Handbook of Pharmaceutical Granulation Technology: Second Edition, edited by Dilip M. Parikh 155. Percutaneous Absorption: Drugs–Cosmetics–Mechanisms–Methodology, Fourth Edition, edited by Robert L. Bronaugh and Howard I. Maibach 156. Pharmacogenomics: Second Edition, edited by Werner Kalow, Urs A. Meyer and Rachel F. Tyndale 157. Pharmaceutical Process Scale-Up, Second Edition, edited by Michael Levin 158. Microencapsulation: Methods and Industrial Applications, Second Edition, edited by Simon Benita 159. Nanoparticle Technology for Drug Delivery, edited by Ram B. Gupta and Uday B. Kompella
  • 9.
  • 10. Nanoparticle Technology for Drug Delivery edited by Ram B. Gupta Auburn University Auburn, Alabama Uday B. Kompella University of Nebraska Medical Center Omaha, Nebraska New York London
  • 11. Published in 2006 by Taylor & Francis Group 270 Madison Avenue New York, NY 10016 © 2006 by Taylor & Francis Group, LLC No claim to original U.S. Government works Printed in the United States of America on acid-free paper 10 9 8 7 6 5 4 3 2 1 International Standard Book Number-10: 1-57444-857-9 (Hardcover) International Standard Book Number-13: 978-1-57444-857-3 (Hardcover) This book contains information obtained from authentic and highly regarded sources. Reprinted material is quoted with permission, and sources are indicated. A wide variety of references are listed. Reasonable efforts have been made to publish reliable data and information, but the author and the publisher cannot assume responsibility for the validity of all materials or for the consequences of their use. No part of this book may be reprinted, reproduced, transmitted, or utilized in any form by any electronic, mechanical, or other means, now known or hereafter invented, including photocopying, microfilming, and recording, or in any information storage or retrieval system, without written permission from the publishers. For permission to photocopy or use material electronically from this work, please access www.copyright.com (http://www.copyright.com/) or contact the Copyright Clearance Center, Inc. (CCC) 222 Rosewood Drive, Danvers, MA 01923, 978-750-8400. CCC is a not-for-profit organization that provides licenses and registration for a variety of users. For organizations that have been granted a photocopy license by the CCC, a separate system of payment has been arranged. Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and are used only for identification and explanation without intent to infringe. Library of Congress Cataloging-in-Publication Data Catalog record is available from the Library of Congress Visit the Taylor & Francis Web site at http://www.taylorandfrancis.com Taylor & Francis Group is the Academic Division of Informa plc.
  • 12. Preface Products of nanotechnology are expected to revolutionize modern medicine, as evidenced by recent scientific advances and global initiatives to support nanotechnology and nano- medicine research. The field of drug delivery is a direct bene- ficiary of these advancements. Due to their versatility in targeting tissues, accessing deep molecular targets, and con- trolling drug release, nanoparticles are helping address chal- lenges to face the delivery of modern, as well as conventional drugs. Since the majority of drug products employ solids, nanoparticles are expected to have a broad impact on drug product development. The purpose of this book is to present practical issues in the manufacturing and biological applica- tion of nanoparticles. Drug delivery scientists in industry, academia, and regulatory agencies, as well as students in bio- medical engineering, chemical engineering, pharmaceutical sciences, and other sciences with an interest in drug delivery, iii
  • 13. will find this book useful. It can also be used as a textbook for drug delivery courses focusing on nanoparticles. This book is organized into four sections. The first section describes the distinguishing fundamental properties of nano- particles (Chap. 1) as well as technologies for nanoparticle manufacturing (Chaps. 2–4). Nanoparticles can be manufac- tured by either breaking macro-particles using technologies such as milling and homogenization (Chap. 2) or by building particles from molecules dissolved in a solution using super- critical fluid technology (Chap. 3). Nanoparticle manufacturing and properties can be further optimized by employing poly- mers or proteins as stabilizers (Chap. 4). The second section describes the characterization of nano- particles at the material or physicochemical level (Chap. 5) and relates these properties to the delivery and effectiveness of nanoparticles (Chap. 6) as well as toxicological characteristics (Chap. 7). The third section presents the various applications of nanoparticles in drug delivery. Depending on the route and purpose of drug delivery, the requirements for nanoparticulate systems can vary. These aspects are discussed in Chapter 8 for injectable delivery, Chapter 9 for oral delivery, Chapter 10 for brain delivery, Chapter 11 for ocular delivery, and Chapter 12 for gene delivery. Finally, the fourth section provides an overview of the clinical, ethical, and regulatory issues of nanoparticle-based drug delivery. These are evolving areas and the drug product development experience with nanoparticles is limited. As more data is gathered on the safety and efficacy of nanoparti- culate systems, a clearer view will emerge. Preparation of this book would not have been possible without the valuable contributions from various experts in the field. We deeply appreciate their timely contributions. Also, we are thankful to our colleagues at Auburn University and the University of Nebraska Medical Center for their support in preparing this book. Ram B. Gupta Uday B. Kompella iv Preface
  • 14. Contents Preface . . . . iii Contributors . . . . xi PART I: TECHNOLOGIES FOR NANOPARTICLE MANUFACTURING 1. Fundamentals of Drug Nanoparticles . . . . . . . . 1 Ram B. Gupta Introduction . . . . 1 Nanoparticle Size . . . . 2 Nanoparticle Surface . . . . 3 Nanoparticle Suspension and Settling . . . . 4 Magnetic and Optical Properties . . . . 6 Production of Nanoparticles . . . . 6 Biological Transport of Nanoparticles . . . . 12 Conclusions . . . . 17 References . . . . 18 v
  • 15. 2. Manufacturing of Nanoparticles by Milling and Homogenization Techniques . . . . . . . . . . . 21 Rainer H. Mu€ller, Jan Mo€schwitzer, and Faris Nadiem Bushrab Introduction . . . . 21 Pearl/Ball-Milling Technology for the Production of Drug Nanocrystals . . . . 25 Drug Nanocrystals Produced by High-Pressure Homogenization . . . . 28 Production of Drug Nanocrystal Compounds by Spray-Drying . . . . 33 Production in Nonaqueous Liquids . . . . 35 Production in Hot-Melted Matrices . . . . 37 Pelletization Techniques . . . . 41 Direct Compress . . . . 45 References . . . . 47 3. Supercritical Fluid Technology for Particle Engineering . . . . . . . . . . . . . . . . . . . . . 53 Ram B. Gupta Introduction . . . . 53 Supercritical CO2 . . . . 54 Solubility in Supercritical CO2 . . . . 55 Rapid Expansion of Supercritical Solution for Particle Formation . . . . 59 RESS with Solid Cosolvent for Nanoparticle Formation . . . . 63 Supercritical Antisolvent Process for Particle Formation . . . . 66 SAS with Enhanced Mass (EM) Transfer (SAS-EM) Process for Nanoparticle Formation . . . . 69 Fundamentals Governing Particle Formation with RESS and SAS . . . . 70 Other Applications of SCFs for Particle Engineering . . . . 74 Safety and Health Issues . . . . 78 Conclusions . . . . 78 References . . . . 79 vi Contents
  • 16. 4. Polymer or Protein Stabilized Nanoparticles from Emulsions . . . . . . . . . . . . . . . . . . . . . . . . . 85 Ram B. Gupta Introduction . . . . 85 Emulsification Solvent Evaporation Process . . . . 86 Emulsification . . . . 87 Nanoparticle Hardening . . . . 93 Residual Solvent and Emulsifier . . . . 97 Protein Stabilized Nanoparticles . . . . 98 Conclusions . . . . 99 References . . . . 101 PART II: NANOPARTICLE CHARACTERIZATION AND PROPERTIES 5. Physical Characterization of Nanoparticles . . . 103 Roy J. Haskell Introduction . . . . 103 Measurement of Size . . . . 105 Available Methods . . . . 109 In Vitro Release . . . . 119 Example: Particle Size . . . . 121 Conclusions . . . . 130 References . . . . 132 6. Nanoparticle Interface: An Important Determinant in Nanoparticle-Mediated Drug/Gene Delivery . . . . . . . . . . . . . . . . . . . . . . 139 Sanjeeb K. Sahoo and Vinod Labhasetwar Introduction . . . . 139 Influence of Emulsifier on Pharmaceutical Properties of Nanoparticles . . . . 140 Implication on Cellular Uptake/Toxicity/Gene Delivery . . . . 148 Biodistribution . . . . 153 Conclusions . . . . 154 References . . . . 154 Contents vii
  • 17. 7. Toxicological Characterization of Engineered Nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . 161 Paul J. A. Borm and Roel P. F. Schins Introduction . . . . 161 Inhalation of Particles . . . . 164 Effects of Nanoparticles . . . . 170 Screening Engineered NP for Toxicological Hazards . . . . 178 Conclusion . . . . 187 References . . . . 188 PART III: DRUG DELIVERY APPLICATIONS OF NANOPARTICLES 8. Injectable Nanoparticles for Efficient Drug Delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 199 Barrett Rabinow and Mahesh V. Chaubal Introduction: Medical Needs Addressable by Nanoparticulate Drug Delivery . . . . 199 Types of Carriers . . . . 209 Coating Functionality . . . . 215 External Assistance in Targeting . . . . 215 Drugs Incorporated . . . . 216 Clinical Development . . . . 217 Conclusions . . . . 220 References . . . . 221 9. Polymeric Nanoparticles for Oral Drug Delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 231 Vivekanand Bhardwaj and Majeti Naga Venkata Ravi Kumar Introduction . . . . 231 Physiology of GIT with Relevance to Particulate Uptake . . . . 232 Particle Size and Surface Charge: Critical Factors in Particle Absorption . . . . 236 Bioadhesion . . . . 237 Tracer Techniques . . . . 241 viii Contents
  • 18. In Vitro and In Vivo Models . . . . 243 Nanoparticle Formulation . . . . 244 Applications . . . . 253 Future Directions . . . . 260 References . . . . 262 10. Brain Delivery by Nanoparticles . . . . . . . . . . . 273 Svetlana Gelperina Introduction . . . . 273 Biodistribution Studies . . . . 276 Pharmacological Activity . . . . 289 Mechanisms of Drug Delivery to the Brain by Means of Polymeric NP . . . . 296 Conclusions . . . . 309 References . . . . 311 11. Nanoparticles for Ocular Drug Delivery . . . . . 319 Aniruddha C. Amrite and Uday B. Kompella Introduction . . . . 319 Disposition of Nanoparticles in the Eye . . . . 322 Ocular Drug Delivery Enhancement Using Nanoparticles . . . . 336 Safety and Tolerability of Particulate Systems . . . . 347 Conclusions . . . . 352 References . . . . 353 12. DNA Nanoparticle Gene Delivery Systems . . . 361 Moses O. Oyewumi and Kevin G. Rice Gene Delivery Vectors . . . . 361 Polymers Used to Prepare DNA Nanoparticles . . . . 363 Physical Properties of DNA Nanoparticles . . . . 365 Biodistribution and Trafficking of DNA Nanoparticles . . . . 371 Conclusions . . . . 372 References . . . . 373 Contents ix
  • 19. PART IV: CLINICAL, ETHICAL, AND REGULATORY ISSUES 13. Nanotechnology and Nanoparticles: Clinical, Ethical, and Regulatory Issues . . . . . . . . . . . . 381 Makena Hammond and Uday B. Kompella Introduction . . . . 381 Clinical Aspects . . . . 382 Environmental, Social, and Ethical Issues . . . . 385 Regulatory Challenges . . . . 388 Conclusions . . . . 392 References . . . . 393 Index . . . . 397 x Contents
  • 20. Contributors Aniruddha C. Amrite Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, U.S.A. Vivekanand Bhardwaj Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Punjab, India Paul J. A. Borm Centre of Expertise in Life Sciences, Zuyd University, Heerlen, The Netherlands Faris Nadiem Bushrab Department of Pharmaceutical Technology, Biotechnology and Quality Management, Freie Universita¨t, Berlin, Germany Mahesh V. Chaubal BioPharma Solutions, Baxter Healthcare, Round Lake, Illinois, U.S.A. xi
  • 21. Svetlana Gelperina Institute of Molecular Medicine, Moscow Sechenov Medical Academy, Moscow, Russia Ram B. Gupta Department of Chemical Engineering, Auburn University, Auburn, Alabama, U.S.A. Makena Hammond College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, and Virginia State University, Petersburg, Virginia, U.S.A. Roy J. Haskell Pfizer Corporation, Michigan Pharmaceutical Sciences, Kalamazoo, Michigan, U.S.A. Uday B. Kompella Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, U.S.A. Majeti Naga Venkata Ravi Kumar Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Punjab, India Vinod Labhasetwar Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, U.S.A. Jan Mo¨schwitzer Department of Pharmaceutical Technology, Biotechnology and Quality Management, Freie Universita¨t, Berlin, Germany Rainer H. Mu¨ ller Department of Pharmaceutical Technology, Biotechnology and Quality Management, Freie Universita¨t, Berlin, Germany Moses O. Oyewumi Division of Medicinal and Natural Products Chemistry, College of Pharmacy, University of Iowa, Iowa City, Iowa, U.S.A. Barrett Rabinow BioPharma Solutions, Baxter Healthcare, Round Lake, Illinois, U.S.A. Kevin G. Rice Division of Medicinal and Natural Products Chemistry, College of Pharmacy, University of Iowa, Iowa City, Iowa, U.S.A. xii Contributors
  • 22. Sanjeeb K. Sahoo Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, U.S.A. Roel P. F. Schins Institut fur Umweltmedizinische Forschung (IUF), University of Du¨sseldorf, Du¨sseldorf, Germany Contributors xiii
  • 23.
  • 24. 1 Fundamentals of Drug Nanoparticles RAM B. GUPTA Department of Chemical Engineering, Auburn University, Auburn, Alabama, U.S.A. INTRODUCTION In pharmaceutics, %90% of all medicines, the active ingredi- ent is in the form of solid particles. With the development in nanotechnology, it is now possible to produce drug nanoparti- cles that can be utilized in a variety of innovative ways. New drug delivery pathways can now be used that can increase drug efficacy and reduce side effects. For example, in 2005, the U.S. Food and Drug Administration approved intra- venously administered 130-nm albumin nanoparticles loaded with paclitaxel (AbraxaneTM ) for cancer therapy, which epito- mizes the new products anticipated based on nanoparticulate systems. The new albumin/paclitaxel–nanoparticle formula- tion offers several advantages including elimination of PART I: TECHNOLOGIES FOR NANOPARTICLE MANUFACTURING 1
  • 25. toxicity because of cremophor, a solvent used in the previous formulation, and improved efficacy due to the greater dose of the drug that can be administered and delivered. For better development of the nanoparticulate systems, it is essential to understand the pharmaceutically relevant properties of nano- particles, which is the purpose of this chapter and this book in general. In the following narrative, some fundamental prop- erties of nanoparticles including their size, surface area, set- tling velocity, magnetic and optical properties, and biological transport are brought into the perspective of drug delivery. NANOPARTICLE SIZE To put the size of nanoparticles in perspective, Table 1 com- pares sizes of various objects. Because of the comparable size of the components in the human cells, nanoparticles are of great interest in drug delivery. It appears that nature, in making the biological systems, has extensively used nan- ometer scale. If one has to go hand in hand with nature in treating the diseases one needs to use the same scale, whether it is correcting a faulty gene, killing leprosy bacteria sitting inside the body cells, blocking the multiplication of viral gen- ome, killing a cancer cell, repairing the cellular metabolism, or preventing wrinkles or other signs of aging. One cannot use a human arm to massage the hurt leg of an ant. Size matching is important in carrying out any activity. Drug delivery is aimed at influencing the biochemistry of the body. Table 1 Typical Size of Various Objects Object Size (nm) Carbon atom 0.1 DNA double helix (diameter) 3 Ribosome 10 Virus 100 Bacterium 1,000 Red blood cell 5,000 Human hair (diameter) 50,000 Resolution of unaided human eyes 100,000 2 Gupta
  • 26. The basic unit of the biological processes is the cell and the biochemical reactions inside it. With the advent of nanoparti- cles it is now possible to selectively influence the cellular pro- cesses at their natural scales. NANOPARTICLE SURFACE We can generally see and discern objects as small as 100,000 nm (100 mm). It is only in the past 300 years, with the invention of microscopes, that we can see smaller objects. Today, one can see objects as small as individual atoms (about 0.1 nm) using the scanning probe microscope. Owing to their small size, nanoparticles exhibit interesting properties, mak- ing them suitable for a variety of drug delivery applications. The number of molecules present on a particle surface increases as the particle size decreases. For a spherical solid particle of diameter d, surface area per unit mass,Sg, is given as Sg ¼ pd2 4 pd3 rs 6 À1 ¼ 3 2drs ð1Þ where rs is the solid density. If the molecular diameter is s, then the percentage of molecules on the surface monolayer is given as %Surface molecules ¼ ð4=3Þp½d3 À ðd À sÞ3 Š ð4=3Þp½d3Š 100 ¼ 100 s d 3 À3 s d 2 þ3 s d ! ð2Þ For a typical low–molecular weight drug molecule of 1-nm diameter, %surface molecules are calculated in Table 2. It is interesting to see that for a 10,000-nm (or 10-mm) particle, a very small percentage of the molecules are present on the surface. Hence, the dissolution rate is much lower for the microparticles when compared to nanoparticles. When the particles are of nanometer length scale, surface irregularities can play an important role in adhesion, as the irregularities may be of the same order as the particles (1). Fundamentals of Drug Nanoparticles 3
  • 27. Nanoparticles can show a strong adhesion because of the increased contact area for van der Waals attraction. For example, Lamprecht et al. (2) observed differential uptake/ adhesion of polystyrene particle to inflamed colonic mucosa, with the deposition 5.2%, 9.1%, and 14.5% for 10-mm, 1000-nm, and 100-nm particles, respectively. NANOPARTICLE SUSPENSION AND SETTLING Because of the small size of the nanoparticles, it is easy to keep them suspended in a liquid. Large microparticles preci- pitate out more easily because of gravitational force, whereas the gravitational force is much smaller on a nanoparticle. Particle settling velocity, v, is given by Stokes’ law as v ¼ d2 gðrs À rlÞ 18ml ð3Þ where g is gravitation acceleration (9.8 m/sec at sea level), rl is liquid density (997 kg/m3 for water at 25 C), ml is viscosity (0.00089 Pa/sec for water at 25 C). For various particles sizes, settling velocities are calculated in Table 3 for a solid density (rs) of 1700 kg/m3 . Thermal (Brownian) fluctuations resist the particle settlement. According to Einstein’s fluctuation–dissipation theory, average Brownian displacement x in time t is given as x ¼ ffiffiffiffiffiffiffiffiffiffiffiffiffi 2kBTt pmd s ð4Þ Table 2 % Surface Molecules in Particles Particle size (nm) Surface molecules (%) 1 100.00 10 27.10 100 2.97 1,000 0.30 10,000 0.03 4 Gupta
  • 28. where kB is the Boltzman constant (1.38 Â 10À23 J/K), and T is temperature in Kelvin. Table 4 shows displacements for particles of varying sizes in water at 25 C. The Brownian motion of a 1000-nm particle due to thermal fluctuation in water is 1716 nm/sec, which is greater than the settling velocity of 430 nm/sec. Hence, particles below 1000 nm in size will not settle merely because of Brownian motion. This imparts an important property to nanoparticles, that they can be easily kept suspended despite high solid density. Large microparti- cles easily settle out from suspension because of gravity, hence such suspensions need to carry a ‘‘shake well before use’’ label. Also, a microparticle suspension cannot be used for injection. For the nanoparticles, the gravitational pull is not stronger than the random thermal motion of the particles. Hence, nano- paticle suspensions do not settle, which provides a long self-life. However, settling can be induced using centrifugation if needed for particle separation. Particle velocity under centri- fugation is given as: v ¼ p 9 xd2 ðrs À rlÞ m rpm 60 2 ð5Þ Table 4 Brownian Motion of the Particles Particle size (nm) Brownian displacement (nm in 1 sec) 1 54,250 10 17,155 100 5,425 1,000 1,716 10,000 543 Table 3 Particle Settling Velocities Particle size (nm) Settling velocity (nm/sec) 1 0.00043 10 0.043 100 4.30 1,000 430 10,000 43,005 Fundamentals of Drug Nanoparticles 5
  • 29. where the centrifugal rotation is rotations per minute (rpm). For various particle sizes, centrifugal velocities calculated for a solid density (rs) of 1700 kg/m3 in water at 0.1 m from the axis of rotation are presented in Table 5. MAGNETIC AND OPTICAL PROPERTIES Small nanoparticles also exhibit unique magnetic and optical properties. For example, ferromagnetic materials become superparamagnetic below about 20 nm, i.e., the particles do not retain the magnetization because of the lack of magnetic domains; however, they do experience force in the magnetic field. Such materials are useful for targeted delivery of drugs and heat. For example, interaction of electromagnetic pulses with nanoparticles can be utilized for enhancement of drug delivery in solid tumors (3). The particles can be attached to antibodies directed against antigens in tumor vasculature and selectively delivered to tumor blood vessel walls. The local heating of the particles by pulsed electromagnetic radia- tion results in perforation of tumor blood vessels, microcon- vection in the interstitium, and perforation of cancer cell membrane, and therefore provides enhanced delivery of drugs from the blood into cancer cells with minimal thermal and mechanical damage to the normal tissues. Gold and silver nanoparticles show size-dependent opti- cal properties (4). The intrinsic color of nanoparticles changes with size because of surface plasmon resonance. Such nano- particles are useful for molecular sensing, diagnostic, and imaging applications. For example, gold nanoparticles can exhibit different colors based on size (Table 6). PRODUCTION OF NANOPARTICLES Although any particle of a size 1-mm diameter is a nanopar- ticle, several national initiatives are encouraging the develop- ment of particles 100 nm as they might exhibit some unique physical properties, and hence potentially different and use- ful biological properties. However, achieving sizes 100 nm 6 Gupta
  • 31. is more readily feasible with hard materials compared to drug and polymer molecules, which are soft materials. For hard materials, such as silica, metal oxides, and diamonds with melting points above 1000 C, nanoparticles in the 1–100 nm size range have been prepared. However, for drugs that are usually soft materials with melting point below 300 C parti- cles in the 1–100 nm size range are more difficult to prepare. For this reason, it is a reasonable goal to aim at 300 nm par- ticles for drug and polymer materials. There are several suc- cess stories for pharmaceutical materials in this size range. Table 6 Size-Dependent Color Variation of Gold Nanoparticles Wavelength for maximum absorption (nm) Nanoparticle size (nm) In water In AOT/water/isooctane (w0 ¼ 10) 9 519 535 20 523 531 30 525 535 40 526 545 52 528 543 59 535 546 79 550 560 100 567 583 Abbreviation: AOT, sodium bis(2-ethyl hexyl)sulfosuccinate. Source: From Ref. 5. Table 7 Number of Molecules in a Spherical Particle Particle diameter Particle volume (mL) Number of molecules 0.58 nm 8.18 Â 10À22 1 1 nm 4.19 Â 10À21 5.05 10 nm 4.19 Â 10À18 5.05 Â 103 100 nm 4.19 Â 10À15 5.05 Â 106 500 nm 5.24 Â 10À13 6.31 Â 108 1 mm 4.19 Â 10À12 5.05 Â 109 5 mm 5.24 Â 10À10 6.31 Â 1011 1 mm 4.19 Â 10À3 5.05 Â 1018 Note: Drug molecular weight ¼ 500 and solid density ¼ 1 g/mL. 8 Gupta
  • 32. Production of nanoparticles of soft materials is much more challenging than that of hard materials because of the high stickiness of the former. The bulk pharmaceuticals are avail- able in solids of large sizes (e.g., 1-mm-diameter powder), which can be often easily solubilized in solvent to obtain molecular size. Hence, there are two extremes of sizes: molecular size (each particle containing one molecule) and large size (e.g., each particle containing of the order of 1018 molecules). For a drug of 500 molecular weight and 1 g/mL solid density, the numbers of molecules in different size particles are given in Table 7. Hence, to obtain nanoparticles in the 50–300 nm range for drug delivery, one requires of the order of 104 –108 mole- cules in each particle. This size has to be achieved from either solution phase (single molecule) or millimeter-size particle (1018 molecules). The first approach is where the particle is broken down to nano size, whereas in the second approach, the particle will be built up from molecules. The two general approaches for the production of drug nanoparticles are sketched in Figure 1. Milling of Large Particles or Breaking-Down Process Comminuting or grinding or milling is the oldest mechanical unit operation for size reduction of solids and for producing large quantities of particulate materials. Here, the material is subjected to stress, which results in the breakage of the par- ticle. Usually, the applied stress is more concentrated on the Figure 1 Schematic of the two general nanoparticle production techniques. Fundamentals of Drug Nanoparticles 9
  • 33. already present cracks in the material, which causes crack propagation leading to fracture. With the decreasing particle size, materials exhibit increasing plastic behavior making it more difficult to break small particles than large particles. For many materials, a limit in the grindability can be reached where subject to further grinding, no decrease in the particle size is observed (6). An empirical index, known as Bond work index (Wi) has been developed, which represents energy required for grinding (7). Wi ¼ 10 d À1=2 product À d À1=2 feed ð6Þ To reduce the size of a 1-mm particle, the energy required in terms of Bond index is given in Table 8 for various product sizes. Hence, it is very energy intensive to go down to nanopar- ticles-size range. Other than size, parameters of importance are: (i) toughness/brittleness (in tough materials, stress causes plastic deformation, whereas in brittle materials cracks are propagated; hence, size reduction of brittle materi- als is easier than for tough materials; sometimes, a material can be cooled to embrittle), and (ii) hardness, abrasiveness, particle shape and structure, heat sensitivity (only about 2% of the applied energy goes to size reduction, the rest is con- verted to heat; hence, heat-sensitive drugs require cooling), and explodability (most pharmaceuticals are organic materi- als; as the size is reduced air combustibility of the material increases, hence proper inerting is needed). Table 8 Energy Need (Bond Work Index) for Reducing Size of 1-mm-Diameter Particles Product diameter (mm) Energy required (Bond work index) 100 0.68 10 2.85 1 9.68 0.5 13.83 0.1 31.31 10 Gupta
  • 34. Most of the pharmaceutical size reduction operations uti- lize high-shear wet milling for the production of nanoparticles. Milling is explained in detail in chapter 2. Typical operation time for the wet milling may be hours to days, hence the drug has to show stability in that time period, otherwise milling cannot be used for unstable drugs. In addition, one has to be aware of contamination due to milling media. Precipitation from Solution or Building-Up Process In this process, a drug is dissolved in a solvent to achieve molecular solution. Then, the nanoparticle precipitate is obtained either by removing the solvent rapidly or by mixing an antisolvent (nonsolvent) to the solution, reducing its solu- bilizing strength. Initially, nuclei are formed, which grow because of condensation and coagulation giving the final par- ticles. If the rate of desolubilization is slow, then sticky nuclei/ particles are formed that have a higher tendency of agglo- meration, giving large-size final particles. For example, if a drug is dissolved in a solvent (e.g., toluene) and then an Figure 2 Variation of the particle size as the antisolvent and its mixing are varied in the solvent–antisolvent precipitation process. Fundamentals of Drug Nanoparticles 11
  • 35. antisolvent (e.g., methanol) is added with mild mixing, one will obtain drug precipitate of typically 1 mm particle size (Fig. 2). To obtain nanoparticles, a high desolubilization rate is needed, or use of a surfactant is required, which can isolate the particles until they are completely dry. Based on these requirements, two general methods for nanoparticle production are available: (i) supercritical fluid process, and (ii) emulsification–diffusion process. In the precipitation pro- cess, one can add compounds (e.g., polymers for controlled release) that will coprecipitate with the drug for smart drug delivery applications. The key aspect of getting nanoparticles of the desired size and size distribution is to control both the rate of antisol- vent action and the particle coagulation. Precipitation-based processes are described in chapters 3 and 4. BIOLOGICAL TRANSPORT OF NANOPARTICLES For drug delivery, most of the sites are accessible through either microcirculation by blood capillaries or pores present at various surfaces and membranes. Most of the apertures, openings, and gates at cellular or subcellular levels are of nanometer size (Table 9); hence, nanoparticles are the most suited to reach the subcellular level. One of the prime require- ments of any delivery system is its ability to move around freely in available avenues and by crossing various barriers that may come in the way. Regarding the human body, the major passages are the blood vessels through which materials are transported in the body. The blood vessels are not left in any organ as an open outlet of the pipe, rather they become thinner and thinner and are finally converted to capillaries through branching and narrowing. These capillaries go to the close vicinity of the individual cells. After reaching their thinnest sizes, the capillaries start merging with each other to form the veins. These veins then take the contents back to the heart for recirculation. Hence, the supply chain in the body is not in the form of a pipe having an open inlet to the organ and outlet away from the organ. Consequently, for 12 Gupta
  • 36. any moiety to remain in the vasculature, it needs to have its one dimension narrower than the cross-sectional diameter of the narrowest capillaries, which is about 2000 nm. Actually, for efficient transport the nanoparticle should be smaller than 300 nm. But, just moving in the vessels does not serve the drug delivery purpose. The delivery system must reach the site at the destination level. This requires crossing of the blood capillary wall to reach the extracellular fluid of the tissue and then again crossing of other cells, if they are in the way, and entering the target cell. These are the major bar- riers in the transit. A nanoparticle has to do a lot during this sojourn of the carrier through the vessels (capillaries) and across the barriers. There are two routes for crossing the blood capillaries and other cell layers, i.e., transcellular and paracellular. In Table 9 Approximate Sizes of Components in a Typical 20-mm Human Tissue Cell Component Size (nm) Ribosomes 25 Golgi vesicles 30–80 Secretary vesicles 100–1000 Glycogen granules 10–40 Lipid droplets 200–5000 Vaults 55 Lysosomes 500–1000 Proteasomes 11 Peroxisomes 500–1000 Mitochondria 500–1000 Superfine filaments 2–4 Microfilaments 5–7 Thick filaments 15 Microtubules 25 Centrioles 150 Nuclear pores 70–90 Nucleosomes 10 Chromatin 1.9 Source: From Refs. 8, 9. Fundamentals of Drug Nanoparticles 13
  • 37. the transcellular route, the particulate system has to enter the cell from one side and exit the cell from the other side to reach the tissue. The particulate system has to survive the intracellular environment to reach the target tissue. The other route is paracellular. In this, the particlulate system is not required to enter the cell; instead, it moves between the cells. These intercellular areas are known as the junctions. Passing through the junctions would obviate destruction of the carrier by the cell system. Paracellular movement of moieties including ions, larger molecules, and leukocytes is controlled by the cytoskeletal association of tight junctions and the adherence junctions called apical junction complex. While tight junctions act as a regulated barrier, the adherence junctions are responsible for the development and stabilization of the tight junctions. Different epithelial and endothelial barriers have different permeabilities mainly because of the differences in the structure and the presence of tight junctions. While epithelia and brain capillary endothe- lium exhibit a high degree of barrier function, the vascular endothelium in other tissues has greater permeability. The tight junctions control the paracellular transport. For exam- ple, diffusion of large molecules may not be feasible, but migration of white cells is allowed. Understanding of this reg- ulation mechanism is important as this might enable us to pave the way for the movement of nanoparticles in the body without actually entering into the unintended cells. As the nanoparticle-based drug delivery is achieved by particle transport, it is important to understand the blood flow rates and volumes of various organs and tissues. Consid- ering the body’s distribution network, the blood vascular sys- tem, the body could be divided into several compartments based on the distributional sequencing and differentiation by the blood vascular system (Table 10). Nanoparticles can have deep access to the human body because of the particle size and control of surface properties. Experiments by Jani et al. (13,14) have elegantly demon- strated the size effect. Polystyrene particles in the size range 50–3000 nm were fed to rats daily for 10 days at a dose of 1.25 mg/kg. The extent of absorption of the 50-nm particles 14 Gupta
  • 38. Table10VolumesandBloodSupplyofDifferentBodyRegions TissueVolume(mL) Volumeofbloodin equilibriumwithtissue(mL) Bloodflow (mL/min)(mL/dLinanorgan/min) Viscera(wellperfused) Adrenals2062100500 Kidneys3007651240410 Thyroid204980400 Graymatter75037160080 Heart30014824080 Othersmallglandsandorgans160508050 Liverwithportalsystem3,900979158041 Whitematter75010016021 Leantissues(moderatelyperfused) Redmarrow1,400741209 Muscle30,0009803.25 Skin(nutritive)3,000983.25 Nonfatsubcutaneous4,80043701.5 Adiposetissues(poorlyperfused) Fattymarrow2,20037602.7 Fat10,0001232002 Others Bonecortex6,400%0%0%0 Arterialblood1,400 Venousblood4,000 Lungparenchymaltissue6001400 Airinlung2.5þhalfthe tidalvolume Note:Cardiacoutput,6480mL/min;dataareofstandardmen:age30–39years,bodyweight70kg,andsurfacearea1.83mÀ2 . Source:FromRefs.10–12. Fundamentals of Drug Nanoparticles 15
  • 39. was 34% and that of the 100-nm particles was 26%. Of the total absorption, about 7% (50 nm) and 4% (100 nm) were accounted for in the liver, spleen, blood, and bone marrow. Particles 100 nm did not reach the bone marrow, and those 300 nm were absent from the blood. Particles were absent in the heart or the lung tissue. The rapid clearance of circulating particles from the bloodstream coupled with their high uptake by liver and spleen can be overcome by reducing the particle size, and by making the particle surface hydrophilic with coatings, such as poloxamers or poloxamines (15). Gaur et al. (16) observed that 100-nm nanoparticles of polyvinylpyrrolidone had a negligible uptake by the macro- phages in liver and spleen, and 5–10% of these nanoparticles remain in circulation even eight hours after intravenous injection. Because of longer residence in the blood, nanoparti- cles have potential therapeutic applications, particularly in cancer; the cytotoxic agents encapsulated in these particles can be targeted to tumors while minimizing the toxicity to the reticuloendothelial system. Desai et al. (17) studied the effect of poly(d,l-lactide-co- glycolide) (PLGA). particle size (100 nm, 500 nm, 1 mm, and 10 mm) on uptake in rat gastrointestinal tissue. The uptake of 100-nm-size particles by the intestinal tissue was 15–250- fold higher compared to the larger-size microparticles. The uptake also depends on the type of tissue (i.e., Peyer’s patch and nonpatch) and the location (i.e., duodenum or ileum). Depending on the particle size, Peyer’s patch tissue had a 2–200-fold higher uptake of particles than the nonpatch tis- sue. The 100-nm particles were diffused throughout the submucosal layers, while the larger-size particles were predo- minantly localized in the epithelial lining of the tissue, because of the microparticle exclusion phenomena in the gastrointestinal mucosal tissue. Hillyer and Albrecht (18) studied the gastrointestinal uptake and subsequent tissue/organ distribution of 4-, 10-, 28-, and 58-nm-diameter metallic colloidal gold particles fol- lowing oral administration to mice. It was found that colloidal gold uptake is dependent on the particle size: smaller particles cross the gastrointestinal tract more readily. Interestingly, 16 Gupta
  • 40. they observed that the particle uptake occurs in the small intestine by persorption through single, degrading enterocytes in the process of being extruded from a villus. Cellular uptake is greater for nanoparticles compared to microparticles. In cultured human retinal pigment epithelial cells, an increase in the mass uptake of particles was observed with decreasing particle size in the range of 20–2000 nm poly- styrene particles (19). Further, no saturable uptake was observed for these particles up to a concentration of 500 mg/ mL. With 20-nm nanoparticles, the uptake by the 1-cm2 cell monolayer was as high as $20%. Because of possible differences in particle uptake, gene expression efficiencies can also be improved with smaller par- ticles. Prabha et al. (20) studied relative transfectivity of 70- and 202-nm-PLGA nanoparticles in cell culture. The smaller particles showed a 27-fold higher transfection than the larger nanoparticles in COS-7 cell line and a fourfold higher trans- fection in HEK-293 cell line. CONCLUSIONS Nanoparticles offer unique properties as compared to micro- or macroparticles. Salient features include the following: Small size. High surface area. Easy to suspend in liquids. Deep access to cells and organelles. Variable optical and magnetic properties. Particles smaller than 200 nm can be easily sterilized by filtration with a 0.22-mm filter. Drugs, being mostly organic compounds, are more sticky in nature as compared to inorganic materials, such as silica or metal oxides. Hence, it is harder to make smaller nanoparti- cles of drugs compared with hard materials. Drug nanoparti- cles can be produced either by milling of macroparticles or by fast precipitation from solutions, as described in the following chapters. Fundamentals of Drug Nanoparticles 17
  • 41. REFERENCES 1. Chow TS. Size-dependent adhesion of nanoparticles on rough substrates. J Phys: Condens Matter 2003; 15(suppl 2):L83–L87. 2. Lamprecht A, Schafer U, Lehr C-M. Size-dependent bioadhe- sion of micro- and nanoparticulate carriers to the inflamed colonic mucosa. Pharm Res 2001; 18(suppl 6):788–793. 3. Barros APH. Synthesis and Agglomeration of Gold Nanoparti- cles in Reverse Micelles. M.S. thesis, University of Puerto Rico, Mayaguez, PR, 2005. 4. Esenaliev RO. Radiation and nanoparticles for enhancement of drug delivery in solid tumors. PCT Int Appl 2000, WO 2000002590. 5. Kelly L, Coronado E, Zhao LL, Schatz GC. The optical proper- ties of metal nanoparticles: the influence of size, shape, and dielectric environment. J Phys Chem B 2003; 107:668–677. 6. Prior M. Size reduction. In: Kirk-Othmer, Othmer DF, eds. Encyclopedia of Chemical Technology. John Wiley Sons, Inc., 2000. 7. Bond FC. Crushing and grinding calculations. Can Min Metal Trans 1954; 57:466–472. 8. Alberts B, Bray D, Lewis J, Raff M, Roberts K, Watson JD. The Molecular Biology of the Cell. 2nd ed. New York: Garland Publishing, Inc., 1989. 9. Benjamin L. Genes V. New York: Oxford University Press, 1995. 10. Oie S, Benet LZ. The effect of route of administration and distribution on drug action. In: Banker GS, Rhodes CT, eds. Modern Pharmaceutics. New York: Marcel Dekker, 1996. 11. Dedrick RL, Bischoff KB. Pharmacokinetics in applications of the artificial kidney. Chem Eng Progr Symp Ser 1968; 64:32–44. 12. Mapleson WW. An electric analogue for uptake and exchange of inert gases and other agents. J Appl Physiol 1963; 18: 197–204. 13. Jani P, Halbert GW, Langridge J, Florence AT. Nanoparticle uptake by the rat gastrointestinal mucosa: quantitation and 18 Gupta
  • 42. particle size dependency. J Pharm Pharmacol 1990; 42(suppl 12): 821–826. 14. Jani P, Halbert GW, Langridge J, Florence AT. The uptake and translocation of latex nanospheres and microspheres after oral administration to rats. J Pharm Pharmacol 1989; 41(suppl 12): 809–812. 15. Rudt S, Muller RH. In vitro phagocytosis assay of nano- and microparticles by chemiluminescence. III. Uptake of differ- ently sized surface-modified particles, and its correlation to particles properties and in vivo distribution. Eur J Pharm Sci 1993; 1:31–39. 16. Gaur U, Sahoo SK, De TK, Ghosh PC, Maitra A, Ghosh PK. Biodistribution of fluoresceinated dextran using novel nano- particles evading reticuloendothelial system. Int J Pharm 2000; 202(suppl 1–2):1–10. 17. Desai MP, Labhasetwar V, Amidon GL, Levy RJ. Gastrointest- inal uptake of biodegradable microparticles: effect of particle size. Pharm Res 1996; 13(suppl 12):1838–1845. 18. Hillyer, JF, Albrecht RM. Gastrointestinal persorption and tis- sue distribution of differently sized colloidal gold nanoparti- cles. J Pharm Sci 2001; 90(suppl 12):1927–1936. 19. Aukunuru JV, Kompella UB. In vitro delivery of nano- and micro-particles to retinal pigment epithelial (RPE) cells. Drug Deliv Technol 2002; 2(suppl 2):50–57. 20. Prabha S, Zhou W-Z, Panyam J, Labhasetwar V. Size- dependency of nanoparticle-mediated gene transfection: stu- dies with fractionated nanoparticles. Int J Pharm 2002; 244(suppl 1–2):105–115. Fundamentals of Drug Nanoparticles 19
  • 43.
  • 44. 2 Manufacturing of Nanoparticles by Milling and Homogenization Techniques RAINER H. MU¨ LLER, JAN MO¨ SCHWITZER, and FARIS NADIEM BUSHRAB Department of Pharmaceutical Technology, Biotechnology and Quality Management, Freie Universita¨t, Berlin, Germany INTRODUCTION The number of newly developed drugs having a poor solubility and thus exhibiting bioavailability problems after oral admini- stration is steadily increasing. Estimates by the pharma- ceutical companies are that about 40% of the drugs in the pipelines are poorly soluble, and as high as 60% of the com- pounds come directly from the synthesis route (1). Therefore, since a number of years the pharmaceutical development is 21
  • 45. focused on formulation approaches to overcome solubility and related bioavailability problems, so that these new compounds are available for clinical use. Often forgotten, the problem of poor solubility arises even before the preclinical phase, which means that when screening new compounds for pharmaco- logical activity a test formulation needs to be able to lead to sufficiently high blood levels. Therefore, there is an urgent need to come up with a smart formulation approach. One has to differentiate between specific and nonspecific formulations for increasing solubility and, subsequently, bio- availability. Specific approaches can only be applied to certain drug molecules, e.g., in case of cyclodextrins (CDs) to mole- cules that fit into the respective CD ring. In the area of CDs, research is focused on CD derivates with higher solubi- lity of the CD itself and simultaneously reduced side effects of these excipients; for example, the recent development of Cap- tisol1 CDs (2,3). On the other hand, the nonspecific formulation approaches are applicable to almost any drug molecule (apart from a few exceptions). Such a nonspecific formulation approach since many years is micronization, which means converting relatively coarse drug particles to micrometer crystals with a mean diameter in the range of approximately 2–5 mm, and a corresponding size distribution approximately between 0.1 and 20 mm (4). Here, the increase in the surface area leads to an increase in the dissolution velo- city. That means micronization is a formulation approach to overcome the bioavailability problems of drugs of the biophar- maceutical specification class II (BSC II). Drugs of class II are sufficiently permeable but the rate limiting step is a too low dissolution velocity (i.e., low solubility in general is correlated with low dissolution velocity, law by Noyes–Whitney). Now- adays however, many of the new compounds are so poorly soluble that micronization is not sufficient to overcome a too low oral bioavailability. Consequently, the next step taken was to move from micronization to nanonization. By going down one more dimension from the microrange to the nanoworld there is a distinct increase in the surface area and related dissolution velocity. For example, when moving from a spherical 50 mm particle to micronized 5 mm particles, 22 Mu€ller et al.
  • 46. the total surface area enlarges by a factor of 10, moving to 500 nm nanocrystals by a factor of 100. However, there is an additional—but often forgotten—effect further increasing the dissolution velocity, that is, the increase in saturation solubi- lity cs when moving to sizes below 1 mm. Because of the strong curvature of the particles, they possess an increased dissolu- tion pressure comparable to the increased vapor pressure of ultrafine aerosol droplets. The theoretical background is pro- vided by the Kelvin equation and the Ostwald–Freundlich equation, which will not be discussed here in detail (5). Accord- ing to the Noyes–Whitney equation the dissolution velocity dc/dt is proportional to the concentration gradient cs À cx (cs— saturation solubility, cx—concentration in surrounding med- ium, bulk concentration). The increases in saturation solubility of nanocrystals reported are by a factor of about 2 to 4–6 [(6,7) and unpublished data]. The increase is even more pronounced when the nanosized drug material is not crystalline but amor- phous. Preparation of amorphous oleanolic acid nanoparticles increased the saturation solubility up to 10-fold in relation to the coarse drug powder (8). Nanonization has the advantage that it practically can be applied to more or less any drug material. In general, even highly water-sensitive drugs can be reduced to drug nanocrystals, even stored in the form of an aqueous nanosuspension (drug nanocrystals dispersed in aqueous surfactant/stabilizer medium). For example, aqueous Paclitaxel nanosuspension proved to be stable over a period of four years stored at 4 C, i.e., more than 99% of the drug was recovered intact (9). On the other hand, aqueous Paclitaxel solution degrades to an extent of 80% within 25 minutes (10). Drug nanocrystals can be produced by bottom-up or top- down technologies. In the case of bottom-up technologies, one starts with the molecules in solution and moves via associ- ation of these molecules to the formation of solid particles, i.e., it is a classical precipitation process (11). To our know- ledge, there is presently no pharmaceutical product on the market based on precipitation technology. There are a num- ber of reasons, discussed in detail in Ref. (12). Briefly, the use of solvents creates additional costs. In addition, a prere- quisite for precipitation is that the drug is at least soluble Manufacturing of Nanoparticles 23
  • 47. in one solvent, and this solvent needs to be miscible with a nonsolvent. Many of the newly developed compounds; how- ever, are poorly soluble in aqueous and simultaneously in nonaqueous media, thus excluding this formulation approach. In the case of top-down technologies, one starts with a coarse material and applies forces to disintegrate into the nanosize range. The diminution technologies can be categorized into two principal classes: 1. Pearl/ball milling. 2. High-pressure homogenization, and other processes. There are two products on the market based on the pearl/ball-millingtechnologybythecompanyNANOSYSTEMS E´ LAN (13). Rapamune1 coated tablet is the more convenient formulation for the patient compared to the drug solution (Rapamune solution). The bioavailability of the tablet is 27% higher than the solution form (14). Rapamune1 was introduced in the market in 2002 by the company Wyeth. The second is Emend1 , introduced in the market in 2003 by the company MSD1 , Sharp Dohme Gmbh. It is a capsule composed of sucrose, microcrystalline cellulose (MCC), hypro- lose, and sodium dodecylsulfate (SDS) (15). Also, the products based on drug nanocrystals produced with high-pressure homogenization are in clinical phases. Therefore, these two technologies are reviewed in this chapter because of their relevance for the pharmaceutical market. Drug nanocrystals are of high relevance to pharmaceuti- cal products; therefore, it is not surprising that most of the research and development are being done in pharmaceutical companies, especially looking at the production process itself. Of course as a consequence, articles published by companies are very low in number to protect internal knowledge; primar- ily, only published patents are accessible. Even less literature is available on how to transfer the drug nanosuspensions to the final products, i.e., tablets, capsules, and pellets for oral administration or aqueous/lyophilized nanosuspensions for intravenous injection. Producing drug nanocrystals is relatively easy compared to the much more sophisticated technology to formulate a final drug dosage form. A final 24 Mu€ller et al.
  • 48. traditional drug dosage form has to be based on patient conve- nience. However, to fully benefit from the special properties of nanocrystals, they need to be released as ultrafine, non- aggregated suspension from the final dosage form. It could be shown that in the case of strong nanocrsytal aggregation, the dissolution velocity is reduced (16). Therefore, the tricky business is how to transfer the drug nanosuspension to dosage forms with optimized release properties. This chapter also describes the production of tablets, capsules, and pellets. PEARL/BALL-MILLING TECHNOLOGY FOR THE PRODUCTION OF DRUG NANOCRYSTALS Traditional equipment used for micronization of drug powders such as rotor–stator colloid mills (Netzsch) or jet mills (Retsch) are of limited use for the production of nano- crystals. For example, jet milling leads to a drug powder with a size range of roughly between 0.1 and 20 mm, containing only a very small fraction of about 10% in the nanometer range (4). However, it could be shown when running a pearl mill over a sufficiently long milling time, that drug nanosus- pensions can be obtained (13,17,18). These mills consist of a milling container filled with fine milling pearls or larger-sized balls. The container can be static and the milling material is moved by a stirrer; alternatively, the complete container is moved in a complex movement leading consequently to move- ment of the milling pearls. There are different milling materials available, tradi- tionally steel, glass, and zircon oxide are used. New materials are special polymers, i.e., hard polystyrene. A problem asso- ciated with the pearl milling technology is the erosion from the milling material during the milling process. Buchmann et al. (19) reported about the formation of glass microparti- cles when using glass as milling material. In general, very few data have been published on contamination of pharma- ceutical drug nanosuspensions by erosion from the milling material. Most data have been given in the discussions after oral presentations; figures from such discussions range Manufacturing of Nanoparticles 25
  • 49. from 0.1 to 70 ppm contamination. Of course it should be noted that the extent of erosion depends on the solid con- centration of the macrosuspension to be processed, the hardness of the drug, and based on this, the required milling time and milling material. Apart from the milling material, the erosion from the container also needs to be considered. For the parts in contact with the product various materials are offered by companies producing pearl mills depending on the area of application, such as technical purposes, food, or the pharmaceutical industry. Normally, product con- tainers are made of steel and can be covered with various materials to fulfill the required quality specifications of the formulation. Surfactants or stabilizers have to be added for the physical stability of the produced nanosuspensions. In the production process the coarse drug powder is dispersed by high-speed stirring in a surfactant/stabilizer solution to yield a macrosuspension. The choice of surfactants and stabilizers depends not only on the properties of the particles to be suspended (e.g., affinity of surfactant/stabilizer to the crystal surface) but also on the physical principles (electrostatic vs. steric stabilization) and the route of administration. In gen- eral, steric stabilization is recommended as the first choice because it is less susceptible to electrolytes in the gut or blood. Electrolytes reduce the zeta potential and subse- quently impair the physical stability, especially of ionic sur- factants. In many cases an optimal approach is the combination of a steric stabilizer with an ionic surfactant, i.e., the combination of steric and electrostatic stabilization. There is a wide choice of various charged surfactants in case of drug nanocrystals for oral administration. Even relatively ‘‘nasty’’ surfactants, such as the membrane damaging SDS, can be used, of course within the concentration accepted for oral administration, e.g., the formulation of Emend (15). SDS as a low molecular weight surfactant diffuses fast to particle surfaces; it has excellent dispersion properties. Adsorption onto the particle surface leads to high zeta poten- tial values providing good physical stabilities. In case of par- enteral drug nanocrystals, the choice is limited; e.g., for 26 Mu€ller et al.
  • 50. intravenous injection, accepted are lecithins, Poloxamer 188, Tween1 80, low molecular weight polyvinylpyrrolidone (PVP), sodium glycocholate (in combination with lecithin). Drug nanocrystal suspensions for parenteral administrations need to be sterile, depending on the administration route and the volume they need to be pyrogene-free. Production of par- enteral drug nanosuspension using pearl mills is much more tedious compared to producing oral drug nanosuspensions. The equipment needs to be sterilized and the product needs to be separated from the milling pearls by a preferentially aseptic separation process. A terminal sterilization by auto- claving is only possible with a number of products (20). The use of an ionic stabilizer such as lecithin is recom- mended when autoclaving nanosuspensions. The autoclaving temperature of 121 C leads to dehydration of steric stabili- zers, which reduces their ability to stabilize the suspensions. Gamma irradiation of nanosuspensions is an alternative, but is less favoured by the pharmaceutical industry due to regu- latory requirements (e.g., proof of absence of toxic radicals, etc.). From the industrial point of view, in many cases a well-documented aseptic production is easier for the produc- tion of formulations for parenteral administration than gamma irradiation. There are a number of pearl mills available on the market, ranging from laboratory-scale to industrial-scale volumes. The ability for large-scale production is an essential prerequisite for the introduction of a product to the market. One advantage of the pearl mills, apart from being low-cost products, is their ability for scaling up. Assuming, for reasons of simplicity, hexagonal packaging of the milling pearls, 76% of the milling chamber volume will be filled by the pearls. In case of a 1000 L mill this corresponds to 760 L milling mate- rial; based on the apparent density of zircon oxide pearls being 3.69 kg/L, this corresponds to 2.8 tons of milling mate- rial. Figure 1 shows the solution for this problem, a pearl mill with an external suspension container. The suspension is con- tinuously pumped through the pearl mill. This approach reduces the weight of the pearl mill itself, but it prolongs the milling times. Manufacturing of Nanoparticles 27
  • 51. DRUG NANOCRYSTALS PRODUCED BY HIGH-PRESSURE HOMOGENIZATION Theoretical Aspects High-pressure homogenization is a technology that has been applied for many years in various areas for the production of emulsions and suspensions. A distinct advantage of this technology is its ease for scaling up, even to very large volumes. High-pressure homogenization is currently used in the food industry, e.g., homogenization of milk. In the Figure 1 DISPERMAT1 SL: schematic view of a bead mill using recirculation method. Source: From Ref. 21. 28 Mu€ller et al.
  • 52. pharmaceutical industry parenteral emulsions are produced by this technology. Commercial products such as Intralipid1 and Lipofundin1 possess a mean droplet diameter in the range of 200–400 nm (photon correlation spectroscopy data) (22). In the mid-1990s of the last century drug nanosuspen- sions produced with high-pressure homogenization were developed (23–27). Typical pressures for the production of drug nanosuspensions are 1000–1500 bar (corresponding to 100–150 Mpa, 14504–21756 psi); the number of required homogenization cycles vary from 10 to 20 depending on the properties of the drug. Most of the homogenizers used are based on the piston-gap principle, an alternative is the jet- stream technology (Fig. 2). The Microfluidizer1 (MicrofluidicsTM Inc., U.S.A.) is based on the jet-stream principle. Two streams of liquid col- lide, diminution of droplets or crystals is achieved mainly by particle collision, but occurrence of cavitation is also consid- ered. The Microfluidizer has also been described for the pro- duction of drug nanosuspensions; however, according to the patent 10–50 cycle passes were required (28). Such a high Figure 2 Basic homogenization principles: piston-gap (left) and jet-stream arrangement (right). In the piston-gap homogenizer the macrosuspension coming from the sample container is forced to pass through a tiny gap (e.g., 10 mm), particle diminution is affected by shear force, cavitation, and impaction. In jet-stream homogeni- zers the collision of two high-velocity streams leads to particle diminution mainly by impact forces. Manufacturing of Nanoparticles 29
  • 53. cycle number is not convenient for the production scale. Based on our own experiences, the Microfluidizer can be used for the production of drug nanosuspensions in the case of soft drugs. In the case of harder drugs, a larger fraction of particles in the micrometer range remain, which do not exhibit the increase in saturation solubility because of their too large size. For many years cavitation was considered as the major force leading to particle diminution in the high-pressure homogenization process. Consequently, most high-pressure homogenization patents in various application areas focus on water as a dispersion medium. In the piston-gap homoge- nizer the liquid is forced through a tiny homogenization gap, typically in the size range of 5–20 mm (depending on the pressure applied and the viscosity of the dispersion med- ium). Using a Micron Lab 40 the suspension is supplied from a metal cylinder by a piston, the cylinder diameter is approxi- mately 3 cm. The suspension is moved by the piston having an applied pressure between 100 and 1500 bar. In principle the piston-gap homogenizer corresponds to a tube system in which the tube diameter narrows from 3 to 5–20 mm. Accor- ding to the Bernoulli equation, the streaming velocity and dynamic pressure increase extremely, the static pressure in the gap falls below the vapor pressure of water at room tem- perature. A liquid boils when its vapor pressure is equal to the static pressure, which means water starts boiling in the gap at room temperature leading to the formation of gas bubbles. The formation of gas bubbles leads to pressure waves disrup- ting oil droplets or disintegrating crystals. When leaving the homogenization gap, the static pressure increases to normal air pressure, which means the water does not boil any more and the gas bubbles collapse. Collapsing of the gas bubbles (implosion) leads again to shock waves contributing to diminution. There are different definitions of cavitation in the literature, describing cavitation either as the formation of gas bubbles in high streaming liquids or as the formation and subsequent implosion of these gas bubbles. At the end of the 1990s it was found that similar efficient particle diminution can be achieved by homogenization in nonaqueous media such as oils and liquid polyethylene glycols 30 Mu€ller et al.
  • 54. (PEGs), which means media with low vapor pressure. In the case of low vapor pressure liquids, the cavitation in the homo- genization gap is distinctly reduced or does not exist at all. Figure 3 shows the change in static pressure when homoge- nizing in water as dispersion medium (left) and in a low-vapor liquid, whereas the static pressure does not fall below the vapor pressure (right). Based on the aforementioned, cavitation does not seem to be essential for a diminution effect. Major forces are droplet or particle collision and the shear forces occurring in this highly turbulent fluid in the gap possessing a high kinetic energy. Homogenization in nonaqueous liquids has advantages for cer- tain pharmaceutical final dosage forms. Preparation of drug nanocrystals in PEGs or oils (e.g., Miglyol 812 or 829) leads to nanosuspensions that can directly be filled into capsules (see the following) (29,30). It is also possible to homogenize in melted nonaqueous matrices, which are solid at room tempera- ture. Solidification of such a matrix leads to a fixation of drug nanocrystals in the solid matrix, thus minimizing or avoiding crystal contact and subsequent crystal fusion/growth. Figure 3 Variation of the static pressure (—) within the homoge- nizing gap. In the case of water the static pressure falls below the vapor pressure (left), whereas in the case of low-vapor media (right) the static pressure stays above the vapor pressure. Manufacturing of Nanoparticles 31
  • 55. As a consequent next step, after homogenization in water (100% water) and homogenization in nonaqueous media (0% water), homogenization was performed in mixtures con- taining different percentages of water (1–99% water). The dis- persion media were water mixed with water-miscible liquids (e.g., alcohols, glycerol). Preparation of drug nanosuspensions in water–ethanol mixtures is favorable for producing dry pro- ducts, because later the spray drying can be performed under milder conditions when using such a mixture. Homogeniza- tion in water–glycerol mixtures (2.25% of water-free glycerol) leads to isotonic drug nanosuspensions for parenteral administration. The laboratory scale homogenizers used by our group are the continuous and batch Micron Lab 40 (APV Systems, Unna, Germany). In the batch version, the batch size is a minimum of 20 mL and a maximum of 40 mL. A minimum of 20 mL is required for the machine to maintain the homoge- nization pressure because smaller volumes cannot be pro- cessed. In the batch Micron Lab 40, the homogenizer is equipped with two product containers having a maximum volume of 1000 mL. Considering the dead volume in the machine, a minimum batch size of about 200 mL is recom- mended. The advantage of the batch Micron Lab 40 is the relatively small batch volume, but unfortunately it is not pro- duced any more. The successor model by the company APV is the APV-1000; however, the minimum batch size for this homogenizer is 150 mL (31). Scaling up to a size suitable for the production of clinical batches was performed using a Lab 60 unit. This homogenizer has a homogenization capacity of 60 L/hr, and can be qualified and validated (32). The com- mercially available Lab 60 was modified by equipping it with 10 L double-walled product containers; processing is possible in a continuous loop mode (2 kg batch) or alternatively in a batch mode (5–10 kg batch). Because of the termination of the production of the Lab 60 an APV 1000 or APV 2000 is recommended for a batch size in this range. Larger-scale machines from APV are the Gaulin 5.5 (160 L homogenization capacity per hour) or the Rannie 55 (600 L homogenization capacity per hour) at a pressure of 1500 bar. 32 Mu€ller et al.
  • 56. Alternative suppliers of piston-gap homogenizers are the companies Avestin1 (33) and GEA Niro Soavi (34). PRODUCTION OF DRUG NANOCRYSTAL COMPOUNDS BY SPRAY-DRYING For the production of tablets, an aqueous nanosuspension can be used as granulation fluid or a dry form of the nanosuspen- sion, powder, or granulate can be employed. Starting from an aqueous macrosuspension containing the original coarse drug powder, surfactant, and water-soluble excipient, the homogenization process can be performed in an easy one step yielding a fine aqueous nanosuspension. In a subsequent step the water has to be removed from the suspension to obtain a dry powder. One method of removing the water from the for- mulation is freeze drying, but it is complex and cost-intensive leading to a highly sensitive product (35,36). Another simple and most suitable method for the industrial production is spray drying. The drug nanosuspension can directly be pro- duced by high-pressure homogenization in aqueous solutions of water-soluble matrix materials, e.g., polymers [PVP, poly- vinylalcohol or long chained PEG, sugars (saccharose, lac- tose), or sugar alcohols (mannitol, sorbitol)]. Afterward the aqueous drug nanosuspension can be spray dried under ade- quate conditions; the resulting dry powder is composed of drug nanocrystals embedded in a water-soluble matrix (37). Figure 4 schematically represents the whole production pro- cess of drug nanocrystal-loaded spray-dried compounds. The loading capacity of the solid powder with drug nano- crystals can be adjusted by varying the concentrations of exci- pient and surfactant in the original aqueous nanosuspension. One aim of a solid nanoparticulate system is releasing the drug nanocrystals after administration in the gastrointestinal tract (GI) as a fine nonaggregated suspension; the other is to increase the physical stability for long-term storage. Contact of the drug nanocrystals is averted by fixation within the matrix. Thereby, the probability of physical instabilities as, e.g., aggregation and ripening are in principle clearly avoided Manufacturing of Nanoparticles 33
  • 57. or minimized to a negligible extent. However, appropriate investigations have shown a relation between the loading capacity of the compounds and the releasing behavior, as well as the storage stability. Exceeding a certain maximum load- ing capacity of the matrix with drug nanocrystals has an increasing negative effect on particle crystal growth and on release as fine dispersion (38). Figure 5 shows the volume distribution of two spray- dried formulations A and B with increasing drug nanocrystal loadings after release in water. The formulation with the highest drug concentration, formulation B, clearly shows a negligible low but detectable aggregated volume fraction Figure 4 Two-step process of the production of drug nanocrystal- loaded compounds: the drug nanosuspension obtained by high- pressure homogenization (Micron Lab 40) is further processed by spray drying using a Mini Bu¨chi. Drug nanocrystals embedded in the matrix are obtained. 34 Mu€ller et al.
  • 58. and consequently, a reduced percentage within the lower nanometer range. PRODUCTION IN NONAQUEOUS LIQUIDS To avoid the removal of water after high-pressure homogeni- zation in aqueous media, homogenization can be performed directly in nonaqueous media. A number of nonaqueous media are suitable as dispersion media for drug nanocrystals. For example, PEG and triglycerides or self-emulsifying drug delivery systems are ideal liquid candidates and are suited Figure 5 LD volume distributions of spray-dried formulations of Tween1 80 stabilized amphotericin B nanocrystals in PVP matrix after redispersion in water. An adequate volume of distilled water (22–23 C) was added to the compounds to obtain a 1% drug nano- suspension after release from the matrix. LD measurements were started after gently stirring until the matrix material was comple- tely dissolved. (ÁÁÁÁ): Formulation A: 15.4% AmphoB, 7.7% Tween 80, 76.9% PVP (Kollidon 25); (—): formulation B: 50.0% AmphoB, 25.0% Tween 80, 25.0% PVP (Kollidon 25); (–): original nanosus- pension, 1% AmB, 0.5% Tween 80. Abbreviation: LD, laser diffrac- tion; PVP, polyvinylpyrrolidone. Manufacturing of Nanoparticles 35
  • 59. for direct filling of hard or soft gelatine capsules (39). The pro- duction process can be easily performed similar to the process in water. Influence of the Dispersion Media Forces caused by shearing, cavitation, and impaction are domi- nant for the diminution of drug particles during the homoge- nizing process. However, the physical properties of the dispersion media/suspension as well as the type of homogeni- zer and geometry of its dissipation zone highly influence these forces and consequently, the diminution. Especially, the visc- osity of the suspension shows significant effect on the proper- ties of the homogenized products. According to the law by Hagen–Poiseuille, flow rate, pres- sure, tube diameter, as well as the viscosity of the streaming suspension are interdependent in laminar flow systems. The Micron Lab 40 works with a constant flow rate. According to the input requirements the homogenization pressure is auto- matically adjusted by the width of the homogenizing gap (40). In this correlation the viscosity of the fluid/suspension can be considered as a determining factor for the width of the gap. Using a Micron Lab 40 simplified a doubling of the width is observed when decoupling the viscosity. Thus, increasing visc- osities alter the flow conditions within the homogenizing region. A decrease in flow velocity and an increase in gap volume clearly influence the homogenization results. Figure 6 shows the calculated width of the homogenization gap of the Micron Lab 40 in dependency on the fluid viscosity. Broadening of the homogenization gap leads to decreased shear forces and kinetic energies of the nanocrystals; con- sequently, the lower forces affect the breaking of the particles. In summary, the grade of particle diminution is deter- mined by the forces acting on each drug particle during the homogenization process and the drug properties (e.g., hard- ness of crystal, number of imperfections in crystal, and per- centage of amorphous fraction) (41). A particle breaks if the acting force is higher than the breakage stress. The max- imal dispersivity of a nanosuspension is reached if further 36 Mu€ller et al.
  • 60. homogenization cycles show no more effect on particle size distribution. In this case, the acting forces are not high enough for inducing particle breakage and further diminution. For the production of drug nanosuspensions in nonaque- ous media the maximal dispersivity has to be investigated for each drug and dispersion medium. In principle, the media with considerably higher viscosities than water require a higher number of homogenization cycles to achieve identical or similar particle sizes and distributions to lower-viscosity media. Figure 7 shows the particle size distribution of Ampho- tericin B nanoparticles, using laser diffraction (LD), after high-pressure homogenization in different dispersion media. PRODUCTION IN HOT-MELTED MATRICES A further possibility for the production of drug nanocrystals in solid matrices is high-pressure homogenization in hot Figure 6 Width of the homogenizing gap as a function of the kine- matic viscosity of the fluid (Micron Lab 40) at a homogenization pressure of 1500 bar. Manufacturing of Nanoparticles 37
  • 61. melts. It offers advantages over production in aqueous solu- tion and subsequent spray drying. The process is completely anhydrous, avoiding possible drug degradation or instabi- lities. The production can directly be performed by hot high- pressure homogenization in melted material (38,42). The homogenizers Micron Lab 40, batch and continuous, were equipped with temperature control jackets placed around the sample/product containers. Working temperatures up to 100 C (heated with water) or higher (heated with silicon oil) can be selected depending on the melting temperature of the used matrix material. For batch operation, solidification has to be averted between each homogenizing cycle. For homogenizers working in the continuous mode, the product containers must be also heated. Figure 8 shows the tempera- ture control devices for the continuous and batch versions of the Micron Lab 40. As the first production step, a presuspension has to be formed consisting of a melted matrix with the addition of the drug powder and surfactant. In the following production Figure 7 LD particle diameters 25–90% of Amphotericin-B nano- crystals after high-pressure homogenization in PEGs of various chain lengths and thus viscosities. Abbreviations: LD, laser diffrac- tion; PEG, polyethylene glycol. 38 Mu€ller et al.
  • 62. step, the hot presuspension can be directly homogenized in the temperature-controlled homogenizer. After reaching the envisaged particle size and size distribution, the suspension can be solidified at room temperature by applying controlled cooling. Figure 9 shows the principle production process of drug nanocrystals in hot melts. Subsequently, the solid nanodispersions obtained can be processed to granulates by milling, for filling capsules or tablet compaction. Alternatively, the hot melt can directly be filled into hard gelatine or hydroxypropyl methylcellulose (HPMC) capsules (Fig. 10). The absence of water during the whole production pro- cess as well as the short processing times and the one-step process to the final product are especially to be noted using the hot melt method. Given these advantages, this techno- logy also has limitations, which have to be compared with the other technologies (e.g., homogenization in water) for the production of solid nanosuspensions. High-pressure homogenization—identical to pearl milling—can only be performed up to a certain drug concentration. Suspensions Figure 8 Micron Lab 40 with temperature control jackets (J): temperature control jacket for the discontinuous Micron Lab 40 (left) around the sample container (S) containing the suspension to be homogenized, for continuous Micron Lab 40 (right) with jack- ets around the sample container and additional jackets around the two 1000 mL product containers (P). Manufacturing of Nanoparticles 39
  • 63. Figure 9 Schematic of the process utilizing melted matrices: the coarse drug material is added to the solid matrix material, which is then melted for dispersing the drug. The nanosuspension is obtained by high-pressure homogenization. Subsequent cooling leads to drug nanocrystals embedded in a solid matrix. Figure 10 Capsules filled with granulated PEG 2000 containing Amphotericin-B nanocrystals (left), tablets produced by compaction of the granulate (right). Abbreviation: PEG, polyethylene glycol. 40 Mu€ller et al.
  • 64. can show paste-like properties at high solids content (e.g., 30% or 40%). The resulting rheological properties, espe- cially high viscosity, lead to suboptimal flow conditions within the homogenization gap. Depending on the homogeni- zer design, some suspensions with higher viscosities can also be processed (e.g., feeding the suspension to the homogenizer by applying air pressure or using a piston, e.g., PANDA range of GEA, Niro-Soavi, Stansted homogenizers) (43). For example, using a Stansted homogenizer a suspension of 40% solid can be processed without any problem (44). How- ever, it should be noted that the viscosity of a suspension does not only depend on the solid content but also on the size, size distribution, and shape of the particles. Depending on these factors particles can form three-dimensional struc- tures in concentrated suspensions with different viscosities. In turn, it is also possible to reduce the viscosity of a highly concentrated suspension by optimizing the size distribution (i.e., making it more polydisperse). PELLETIZATION TECHNIQUES Introduction The nanonization of drugs results in general in a liquid product from most of the techniques described in this chapter. These nanosuspensions have shown excellent long-term stability without Ostwald ripening or chemical alteration (9,45). In some special cases, the nanosuspension can be directly used as a final product, for instance, as pediatric or geriatric dosage forms if the drug absorption rate is limited only by the solubi- lity and dissolution rate of the drug. Apart from this—in case of oral administration—a dry dosage form is clearly preferred for the reasons of convenience (i.e., marketing aspects). There are also other cases in which a more sophisticated dosage form is needed, e.g., to prevent the drug from degradation, to achieve a controlled drug release or to enable better drug targeting. For these reasons, the nanosuspension can be transformed into a solid dosage form by using established techniques, like pelleti- zation, granulation, spray drying, or lyophilization. Manufacturing of Nanoparticles 41
  • 65. Many different pelletization techniques are known, but the most commonly used techniques are the extrusion–spher- onization and the drug layering onto sugar spheres. The choice of the pellet type depends on the required drug content, the drug properties, and the available technical equipment. Irre- spective of the pelletization technique applied, a multiparticu- late dosage form with distinct advantages in comparison to single-unit dosage forms will be obtained. Multiparticulate dosage forms, such as coated pellet systems, show a faster and more predictable gastric emptying and more uniform drug distribution in the GI tract with less inter- and intraindividual variability in bioavailability (46). A broad distribution of the pellets in the gut lumen can enhance the complete redisper- sion of the nanoparticles from the final solid dosage form. The incorporation of drug nanocrystals in the various pellet systems is schematically shown in Figure 11. Matrix Pellet Preparation Aqueous nanosuspensions can be mixed with matrix materials (fillers such as MCC, lactose, or starch); in addition, the nanosuspension works as a binder and wetting fluid for the Figure 11 Schematic drawing of different pellet types containing drug nanocrystals: coated pellet with a sugar bead as core material and a compact layer consisting of a binder/drug nanocrystal layer (left) and a coated matrix pellet with a matrix consisting of a binder/drug nanocrystal mixture as core material (right). 42 Mu€ller et al.
  • 66. extrusion process (27,47–49). Binders like gelatine, HPMC, chitosans, or other polymers can be added to the nanosus- pension before the high-pressure homogenization, which sim- plifies the production process. Alternatively, they can be dispersed in the produced nanosuspension after the high- pressure homogenization. On the one hand these binders are necessary for the extrusion process, but on the other they can also positively influence the properties of the nanosuspen- sion or the nanoparticles. The increased viscosity of the nano- suspension leads to an increased physical stability of the nanosuspension with a decreased tendency of sedimentation, an important factor to obtain reproducible drug content in the final product. Another important point is the possibility to change the zeta potential of the drug nanocrystals by using charged polymers (i.e., chitosan or alginate) to increase the nanosuspension stability under the GI conditions and to achieve better drug targeting (50–53). If the nanosuspension is used as described earlier, one limitation is the maximum achievable drug content of the final product. In order to overcome this problem an additional drying step, such as spray drying, has to be performed to obtain a fine nanocrystalline powder. This powder can be admixed to the matrix material to obtain a mass highly loaded with drug nanocrystals and ready for the extrusion and subsequent spheronization. Afterward a coating can be applied to the matrix cores to modify their drug release properties. Figure 12 shows the major steps in the production of matrix pellets containing drug nanocrystals. A detailed view on these pellets is given in Figure 13. Pellet Preparation by Nanosuspension Layering An alternative way to transfer a prior produced nanosuspen- sion into a pellet formulation is the suspension layering onto sugar spheres (54). The binders that are necessary for this process can also be added before the high-pressure homogeni- zation process resulting in the improved nanosuspension properties mentioned earlier. A schematic production process is shown in Figure 14. Manufacturing of Nanoparticles 43
  • 67. The most important difference between the matrix cores and layered cores is the different drug loading. For the pro- duction of matrix cores from an aqueous nanosuspension without an additional drying step, the drug loading is limited to 4.5%, based on Equation (1), whereas the drug loading of layered cores can be raised by increasing the layering level almost without any limitations. (J. Moschwitzer and R. H. Muller, submitted for publication.) Drug contentðMCÞ ¼ 30% drug contentðNÞÂ15gN 100gðtotal pellet massÞ ¼ 4:5% ð1Þ Calculated maximal achievable drug content in matrix cores without additional drying steps: MC ¼ matrix core, N ¼ nano- suspension (30% is an achievable drug concentration in the nanosuspension, of course depending on the formulation and equipment). Figure 12 Production of drug nanocrystal-loaded matrix cores: the drug nanosuspension obtained by high-pressure homogeniza- tion (Micron Lab 40) is admixed to the matrix material, pellets are prepared by extrusion–spheronization and can be subsequently coated with polymers with the same equipment to modify the drug release properties. 44 Mu€ller et al.