In the 1960s and 1970s, appli-cation of the physical sciences to the problem of oral drug delivery produced the first wave of major advances that shaped the development of the modern com
Trang 3Thomas J Long School of Pharmacy and Health Sciences
University of the Pacific
A JOHN WILEY & SONS, INC., PUBLICATION
Trang 4Copyright © 2011 John Wiley & Sons, Inc All rights reserved.
Published by John Wiley & Sons, Inc., Hoboken, New Jersey
Published simultaneously in Canada
No part of this publication may be reproduced, stored in a retrieval system, or transmitted in any form or by any means, electronic, mechanical, photocopying, recording, scanning, or otherwise, except as permitted under Section 107 or 108 of the 1976 United States Copyright Act, without either the prior written permission of the Publisher, or authorization through payment of the appropriate per-copy fee to the Copyright Clearance Center, Inc.,
222 Rosewood Drive, Danvers, MA 01923, (978) 750-8400, fax (978) 750-4470, or on the web at www.copyright.com Requests to the Publisher for permission should be addressed to the Permissions Department, John Wiley & Sons, Inc., 111 River Street, Hoboken, NJ 07030, (201) 748-6011, fax (201) 748-6008, or online at http://www.wiley.com/go/permission.
Limit of Liability/Disclaimer of Warranty: While the publisher and author have used their best efforts in preparing this book, they make no representations
or warranties with respect to the accuracy or completeness of the contents of this book and specifically disclaim any implied warranties of merchantability
or fitness for a particular purpose No warranty may be created or extended by sales representatives or written sales materials The advice and strategies contained herein may not be suitable for your situation You should consult with a professional where appropriate Neither the publisher nor author shall
be liable for any loss of profit or any other commercial damages, including but not limited to special, incidental, consequential, or other damages For general information on our other products and services or for technical support, please contact our Customer Care Department within the United States at (800) 762-2974, outside the United States at (317) 572-3993 or fax (317) 572-4002.
Wiley also publishes its books in a variety of electronic formats Some content that appears in print may not be available in electronic formats For more information about Wiley products, visit our web site at www.wiley.com.
Library of Congress Cataloging-in-Publication Data:
Oral bioavailability : basic principles, advanced concepts, and applications / edited by Ming Hu, Xiaoling Li.
p ; cm – (Wiley series in drug discovery and development)
Includes bibliographical references.
ISBN 978-0-470-26099-9 (cloth)
1 Drugs–Bioavailability 2 Drug development 3 Intestinal absorption I Hu, Ming, Ph D.
II Li, Xiaoling, Ph.D III Series: Wiley series in drug discovery and development.
[DNLM: 1 Biological Availability 2 Drug Delivery Systems 3 Intestinal Absorption QV 38]
RM301.6.O73 2011
615 .19– dc22
2011002983 oBook ISBN: 978-1-118-06759-8
ePDF ISBN: 978-1-118-06752-9
ePub ISBN: 978-1-118-06758-1
10 9 8 7 6 5 4 3 2 1
Trang 5to my mom Qihua Chang whose constant love and encouragement persists to this date,
to my wife Yanping Wang whose company endears constant push for perfection, and
to my children Vivian and William whose energy and noise are missed now they are in college.
—Ming Hu Dedicated to my grandmother Yunzhi Su,
my parents Bailing Li and Jie Hu,
my wife Xinghang, and
my children Richard and Louis for their unconditional love, encouragement, and understanding.
—Xiaoling Li
Trang 6Ming Hu and Xiaoling Li
2 Physicochemical Characterization of Pharmaceutical Solids 7
Smita Debnath
Lauren Wiser, Xiaoling Gao, Bhaskara Jasti, and Xiaoling Li
4 In Vitro Dissolution of Pharmaceutical Solids 39
Josephine L P Soh and Paul W S Heng
5 Biological and Physiological Features of the Gastrointestinal
Paul C Ho
6 Absorption of Drugs via Passive Diffusion and Carrier-Mediated
Miki Susanto Park and Jae H Chang
7 In Vitro–In Vivo Correlations of Pharmaceutical Dosage Forms 77
Deliang Zhou and Yihong Qiu
Rashim Singh and Ming Hu
9 Efflux of Drugs via Transporters —The Antiabsorption Pathway 111
Jae H Chang, James A Uchizono, and Miki Susanto Park
Leslie M Tompkins and Hongbing Wang
vii
Trang 711 Protein Binding of Drugs 145
Antonia Kotsiou and Christine Tesseromatis
12 Urinary Excretion of Drugs and Drug Reabsorption 167
Pankaj Gupta, Bo Feng, and Jack Cook
13 Pharmacokinetic Behaviors of Orally Administered Drugs 183
Jaime A Y´a˜nez, Dion R Brocks, Laird M Forrest, and Neal M Davies
Venugopal P Marasanapalle, Xiaoling Li, and Bhaskara R Jasti
15 Drug–Drug Interactions and Drug–Dietary Chemical Interactions 233
Ge Lin, Zhong Zuo, Na Li, and Li Zhang
16 Anatomical and Physiological Factors Affecting Oral Drug
Ayman El-Kattan, Susan Hurst, Joanne Brodfuehrer, and Cho-Ming Loi
Zhong Qiu Liu and Ming Hu
18 Drug Transporters and Their Role in Absorption and Disposition
David J Lindley, Stephen M Carl, Dea Herrera-Ruiz, Li F Pan, Lori B Karpes,
Jonathan M E Goole, Olafur S Gudmundsson, and Gregory T Knipp
Takashi Sekine and Hiroyuki Kusuhara
John R Cardinal and Avinash Nangia
21 Lipid-Based and Self-Emulsifying Oral Drug Delivery Systems 343
Sravan Penchala, Anh-Nhan Pham, Ying Huang, and Jeffrey Wang
22 Prodrug Strategies to Enhance Oral Drug Absorption 355
Sai H S Boddu, Deep Kwatra, and Ashim K Mitra
Puchun Liu and Steven Dinh
Marilyn E Morris and Yash A Gandhi
25 Interplay Between Efflux Transporters and Metabolic Enzymes 401
Stephen Wang
26 Regulatory Considerations in Metabolism- and Transporter-Based Drug
Yuanchao (Derek) Zhang, Lei Zhang, John M Strong, and Shiew-Mei Huang
27 Caco-2 Cell Culture Model for Oral Drug Absorption 431
Kaustubh Kulkarni and Ming Hu
Trang 8CONTENTS ix
28 MDCK Cells and Other Cell-Culture Models of Oral Drug Absorption 443
Deep Kwatra, Sai H S Boddu, and Ashim K Mitra
29 Intestinal Perfusion Methods for Oral Drug Absorptions 461
Wei Zhu and Eun Ju Jeong
30 Liver Perfusion and Primary Hepatocytes for Studying
Cindy Q Xia, Chuang Lu, and Suresh K Balani
31 In vivo Methods for Oral Bioavailability Studies 493
Ana Ruiz-Garcia and Marival Bermejo
32 Determination of Regulation of Drug-Metabolizing Enzymes and
Bin Zhang and Wen Xie
33 Computational and Pharmacoinformatic Approaches to Oral
Miguel ´ Angel Cabrera-P´erez and Isabel Gonz´alez- ´ Alvarez
Trang 9In Spring of 1983, I took a position at The University
of Michigan There I met my first Chinese student, Ming
Hu, from mainland China, and began a personal and
professional relationship that has lasted for nearly 30 years
He is now a Professor at the University of Houston and
one of the two editors of this book I am very pleased to
have observed his contributions to science and his success
as a scientist over the nearly 30 years I have known him
and followed his career It is a pleasure to write this
foreword for this book coedited by Ming and his former
classmate at Shanghai Medical University, Prof Xiaoling
Li at University of the Pacific
This book has two purposes, to give readers a
contem-porary understanding of the science of oral bioavailability
and to present the state-of-the-art tools that can be used
to advance the science of oral bioavailability and solve
problems in the development of drug products for oral
administration It presents the advances in the science of
oral bioavailability over the last five decades This
mul-tidisciplinary scientific field has steadily progressed from
an emphasis on physical sciences such as solubility and
solid state properties, to incorporating the significant recent
advances in the biological sciences that emphasize
trans-porters, enzymes, and the biological and physiological
pro-cesses that influence their expression and function
I will note some of the evolutionary and perhaps
revo-lutionary steps this field of oral bioavailability has taken
over last five decades In the 1960s and 1970s,
appli-cation of the physical sciences to the problem of oral
drug delivery produced the first wave of major advances
that shaped the development of the modern commercial
oral dosage form and the science of oral bioavailability
Important physicochemical principles and strategies such
as manipulation of dissolution via physical manipulation
of the drug and drug product and chemical modificationusing prodrugs were developed These approaches are rou-tinely considered and applied in the drug product devel-opment process today The principles governing sustainedand controlled release formulations were developed in those
“early” years (e.g., Higuchi equation), and have becomewidely applied in the later decades of the twentieth cen-tury In the 1980s, important progress in the science oforal bioavailability was led by the development of twocritical absorption models, rat intestinal segment perfu-sion model (developed in my laboratory) and Caco-2 cellmono-layer culture model (developed in Dr Ronald T Bor-chardt’s lab) Prof Hu studied in both laboratories, and was
an early contributor to the development of both of thesesystems for the study of oral absorption These methodshave since become widely adapted by the pharmaceuticalindustries This set the basis for predicting oral absorptionand partitioning bioavailability into its component process,dissolution/release, transport/permeation, and metabolism,notability distinguishing absorption and systemic availabil-ity During the 1980s, major advances were also made
in the study of metabolism in the intestine as well as
the liver, particularly the cytochrome P 450s and resultant
potential drug– drug interaction mechanisms In addition topredicting oral absorption, my laboratory also pioneeredthe concept of exploiting the intestinal mucosal cell pep-tide transporter (hPEPT1) to improve the oral absorption ofpolar drugs by making a prodrug, chemically combining thedrug and an amino acid with a peptide-bond like structure.This mechanistic concept is the basis for the absorption
of many polar drugs and prodrugs The development ofseveral approved prodrugs including valacyclovir and val-ganciclovir, while originally empirical, is based on these
xi
Trang 10xii FOREWORD
transport mechanisms In the 1990s, I established the
con-cept of the Biopharmaceutical Classification System (BCS),
partitioning drugs into classes for drug development and
drug product regulation This BCS approach has found wide
use in drug discovery, development as well as regulation
It has been adapted by regulatory authorities and
govern-ments around the world as a basis for the regulation of drug
product quality
During this same period, the US Food and Drug
Administration began the mandate of requiring studies that
predict drug–drug interactions based on the sciences that
were developed during the past two decades Study of efflux
transporters began in the 1990s and has exploded in the
twenty-first century While efforts to make an inhibitor of
p-glycoprotein for anticancer application have not produced
an approved drug, it is likely that the future will see such
a development The explosion in the study of transporters
is ongoing, with the recent addition of efflux transporters
such as multidrug resistance-related proteins (MRPs), breast
cancer resistant protein (BCRP), and uptake transporters
such as organic anion transporting peptides (OATP), organic
anion transporters (OATs), and carboxylic acid transporter
(CAT) Such advances in our mechanistic understanding
of oral bioavailability will most certainly lead to future
advances in therapy
The advances in the science of oral bioavailability is
driven by the needs to develop orally administered drugs,
which remains the most acceptable patient compliant means
of administering drugs to patients across the globe today
Although the scientific basis was most often the pursuit
of industrial scientists, a lack of rapid advancement in
the science of oral bioavailability became recognized as
a hurdle in the drug development process in the early1990s as many highly potent compounds (high affinity
ligands), for example, HIV in vitro were inactive in humans.
In a timely or even a watershed event, the NationalInstitute of Health in 1994 organized a conference on “OralBioavailability,” where scientists of various backgroundswere organized to address the complex problem facingpotent yet poorly bioavailable drug candidates, particularlyanti-HIV candidates Senior managements in many ofthe major pharmaceutical companies became aware ofand recognized the importance of “bioavailability” as thepharmaceutical industry was working hard to fast track thedevelopment of anti-HIV drugs This led to investment
by the pharmaceutical industries in the technology andscientists to tackle this oral delivery problem While actualnumbers can be hard to obtain and interpret, my impression
is that the attention to bioavailability has led to thedecrease in the percentage of clinical trial failures due tooral bioavailability problems Looking even further intothe future, I believe the science of oral bioavailabilitywill be driven by the needs for personalized medicine,individualized treatment plan tailored to patients, and
by the commercial need to increase the efficiency andefficacy of oral drug product development This bookprovides a comprehensive survey of the modern study
of the science of oral bioavailability in the twenty-firstcentury
GORDON L AMIDON, Ph.D
The University of Michigan, Ann Arbor, MI
Trang 11Since the concept of bioavailability has been introduced,
significant progress has been made in understanding the
science of oral bioavailability and in improving the oral
delivery of drugs Yet, we also find that there is still
much to be discovered to have a good handle on oral
bioavailability As a subject, bioavailability encompasses
the knowledge and technologies from various disciplines
A pharmaceutical scientist in a specific research area will
benefit from a treatise on the topic Hence, the objective
of this book is to provide the framework for fundamental
concepts and contemporary practice of bioavailability in
pharmaceutical research and drug development
It is our belief that this book provides both the basic
concepts to a novice and the advanced knowledge to
veteran pharmaceutical scientists and graduate students
in related research fields Chapter 1 gives a high level
summary of this book The basic concepts of bioavailability
are covered in Chapter 2–13 From Chapter 14 to 26,
the advanced concepts of bioavailability are discussed
in greater depth Various approaches and methods for
improving and studying bioavailability are highlighted in
Chapter 27 to 33 The comprehensive coverage of topics
on bioavailability in this book offers readers a choice of
logically building their knowledge on bioavailability from
basic concepts to advanced applications or `a la carte based
on their specific needs
A book with such diverse contents requires a ciplinary effort Without the efforts of contributors fromdifferent areas, this book would have not been a reality
multidis-We would like to personally thank all authors for theircontributions and patience during the completion of thisbook project Sincere thanks are gratefully extended to MrJonathan Rose at John Wiley and Sons, Inc and Dr BingheWang (the book series editor) for their patience, under-standing, support, and confidence in us We would alsolike to express our appreciations to Mrs Kathy Kassab forher invaluable secretarial assistance, and to Haseen Khanfor her tireless effort in the book production Finally, wewould like to thank the world renowned scientist and lead-ing expert in bioavailability, Prof Gordon L Amidon forwriting an insightful and inspiring forward for this book
Trang 12Suresh K Balani, Drug Metabolism and
Pharmacokinet-ics, Millennium Pharmaceuticals, Inc., 35 Landsdowne
Street, Cambridge, MA 02139
Marival Bermejo, Department of Engineering, Pharmacy
and Pharmaceutical Technology Section, School of
Pharmacy, Universidad Miguel Hern´andez de Elche,
Carretera Alicante Valencia km 87, San Juan de Alicante
03550, Alicante, Spain
Sai H.S Boddu, Division of Pharmaceutical Sciences,
University of Toledo, Toledo, OH
Dion R Brocks, Faculty of Pharmacy, University of
Alberta, Alberta, Canada
Joanne Brodfuehrer, Department of Pharmacokinetics,
Dynamics and Metabolism, Pfizer Global Research and
Development, Cambridge, MA
Miguel Angel ´ Cabrera-P´erez, Molecular Simulation
and Drug Design Department, Centro de Bioactivos
Qu´ımicos, Universidad Central “Marta Abreu” de Las
Villas, Carretera a Camajuan´ı, Km 51/2, Santa Clara,
Villa Clara, C.P 54830, Cuba
John R Cardinal, J R Cardinal Consulting LLC,
Wilm-ington, NC
Stephen M Carl, Department Industrial and Physical
Pharmacy, College of Pharmacy, Nursing and Health
Sciences, Purdue University, West Lafayette, IN 47907
Jae H Chang, Department of Drug Metabolism and
Pharmacokinetics, Genentech, South San Francisco, CA
Jack Cook, Clinical Pharmacology, Specialty Care
Busi-ness Unit, Pfizer Inc., Groton, CT
Neal M Davies, Department of Pharmaceutical Sciences,
College of Pharmacy, Washington State University,Pullman, WA
Smita Debnath, Merck Frosst Canada Ltd, Kirkland,
Bo Feng, Pharmacokinetics, Dynamics and Metabolism,
Pfizer Inc., Groton, CT
Laird M Forrest, Department of Pharmaceutical
Chem-istry, University of Kansas, Lawrence, KS
Yash A Gandhi, Department of Pharmaceutical Sciences,
School of Pharmacy and Pharmaceutical Sciences,University at Buffalo, State University of New York,Buffalo, NY
Xiaoling Gao, Department of Pharmaceutics and
Medici-nal Chemistry, Thomas J Long School of Pharmacy andHealth Sciences, University of the Pacific, Stockton, CA95211
Current Affiliation: Institute of Medical Sciences, hai Jiaotong University School of Medicine, Shanghai,
Shang-PR China
Isabel Gonz´alez- ´ Alvarez, Department of Engineering:
Pharmacy and Pharmaceutical Technology section,School of Pharmacy, Universidad Miguel Hern´andez deElche, Carretera Alicante Valencia km 87., San Juan
03550, Alicante, Spain
xv
Trang 13Jonathan M.E Goole, Laboratory of Pharmaceutics and
Biopharmaceutics, Universite Libre de Bruxelles,
Insti-tute of Pharmacy, 1050 Brussels, Beligum
Olafur S Gudmundsson, Discovery Pharmaceutics,
Pharmaceutical Candidate Optimization, Bristol-Myers
Squibb, Princeton, NJ
Pankaj Gupta, Clinical Pharmacology, Specialty Care
Business Unit, Pfizer Inc., Groton, CT
Paul W.S Heng, Department of Pharmacy, National
University of Singapore, Singapore
Dea Herrera-Ruiz, Universidad Aut´onoma del Estado de
Morelos, Facultad de Farmacia, Cuernavaca, Mexico
Paul C Ho, Department of Pharmacy, National University
of Singapore, Singapore
Ming Hu, Department of Pharmacological and
Pharma-ceutical Sciences, College of Pharmacy, University of
Houston, 1441 Moursund Street, Houston, TX 77030
Shiew-Mei Huang, Offices of Clinical Pharmacology,
Center for Drug Evaluation and Research, Food and
Drug Administration, Building 51, Room 3106 10903
New Hampshire Avenue, Silver Spring, MD 20993
Ying Huang, Department of Pharmaceutical Sciences,
College of Pharmacy, Western University of Health
Sciences, Pomona, CA 91766
Susan Hurst, Department of Pharmacokinetics, Dynamics
and Metabolism, Pfizer Global Research and
Develop-ment, Groton, CT
Bhaskara R Jasti, Department of Pharmaceutics and
Medicinal Chemistry, Thomas J Long School of
Phar-macy and Health Sciences, University of the Pacific,
Stockton, CA 95211
Eun Ju, Korea Institute of Toxicology (KIT), 19
Sin-seongno, Yuseong, Daejeon, 305–343, Republic of
Korea
Gregory T Knipp, Department Industrial and Physical
Pharmacy, College of Pharmacy, Nursing and Health
Sciences, Purdue University, 575 Stadium Mall Dr.,
Room 308A, West Lafayette, IN 47907–2091
Antonia Kotsiou, Department of Pharmacology,
Are-taieion University Hospital, Vas Sophias 76, 11528,
Athens, Greece
Kaustubh Kulkarni, Department of Pharmacological and
Pharmaceutical Sciences, College of Pharmacy,
Univer-sity of Houston, 1441 Moursund Street, Houston, TX
77030
Hiroyuki Kusuhara, Laboratory of Molecular
Pharma-cokinetics, Graduate School of Pharmaceutical Sciences,
The University of Tokyo, Tokyo, Japan
Deep Kwatra, Division of Pharmaceutical Sciences,
School of Pharmacy, University of Missouri-KansasCity, 2464 Charlotte Street, 5005 Rockhill Road, KansasCity, MO 64108-2718
Na Li, Department of Pharmacology, The Chinese
Univer-sity of Hong Kong, Hong Kong
Xiaoling Li, Department of Pharmaceutics and Medicinal
Chemistry, Thomas J Long School of Pharmacy andHealth Sciences, University of the Pacific, Stockton, CA95211
Ge Lin, School of Biomedical Sciences, Faculty of
Medicine, The Chinese University of Hong Kong, HongKong
David J Lindley, Department Industrial and Physical
Pharmacy, College of Pharmacy, Nursing and HealthSciences, Purdue University, West Lafayette, IN 47907
Puchun Liu, Noven Pharmaceuticals, Inc., 11960 SW 144
Street, Miami, FL 33186
Zhong Qiu Liu, Department of Pharmaceutics, School of
Pharmaceutical Sciences, Southern Medical University,Guangzhou 510515, China
Cho-Ming Loi, Department of Pharmacokinetics,
Dynam-ics and Metabolism, Pfizer Global Research and opment, San Diego, CA
Devel-Chuang Lu, Drug Metabolism and Pharmacokinetics,
Mil-lennium Pharmaceuticals, Inc., 35 Landsdowne Street,Cambridge, MA 02139
Venugopal P Marasanapalle, Department of
Pharmaceu-tics and Medicinal Chemistry, Thomas J Long School ofPharmacy and Health Sciences, University of the Pacific,Stockton, CA 95211
Current Affiliation: Forest Research Institute, 220 SeaLane, Farmingdale, NY 11735
Ashim K Mitra, Division of Pharmaceutical Sciences,
School of Pharmacy, University of Missouri-KansasCity, 2464 Charlotte Street, 5005 Rockhill Road, KansasCity, MO 64108-2718
Marilyn E Morris, Department of Pharmaceutical
Sci-ences, School of Pharmacy and Pharmaceutical SciSci-ences,University at Buffalo, State University of New York,Buffalo, New York, NY
Avinash Nangia, Vaunnex Inc., Sharon, Massachusetts
Li F Pan, Department Industrial and Physical Pharmacy,
College of Pharmacy, Nursing and Health Sciences,Purdue University, West Lafayette, IN 47907
Trang 14CONTRIBUTORS xvii
Miki Susanto Park, Department of Pharmaceutics and
Medicinal Chemistry, Thomas J Long School of
Phar-macy and Health Sciences, University of the Pacific,
Stockton, CA 95211
Sravan Penchala, Department of Pharmaceutical
Sci-ences, College of Pharmacy, Western University of
Health Sciences, Pomona, CA 91766
Anh-Nhan Pham, Department of Pharmaceutical
Sci-ences, College of Pharmacy, Western University of
Health Sciences, Pomona, CA 91766
Yihong Qiu, Global Pharmaceutical Regulatory Affairs,
Abbott Laboratories, 200 Abbott Park Rd, RA71-Bldg
AP-30-1, Abbott Park, IL, 60064–6157
Ana Ruiz-Garcia, Clinical Pharmacology, Oncology
Divi-sion, Pfizer Inc, 10646 Science Center Dr CB-10, San
Diego, CA 92121
Takashi Sekine, Department of Pediatrics, Toho
Univer-sity School of Medicine, Tokyo, Japan
Rashim Singh, Department of Pharmacological and
Phar-maceutical Sciences, College of Pharmacy, University
of Houston, 1441 Moursund Street, Houston, TX
Josephine L.P Soh, Pfizer Global Research and
Develop-ment, UK
John M Strong,∗ Offices of Pharmaceutical Sciences,
Center for Drug Evaluation and Research, Food and
Drug Administration, Building 51, Room 3106 10903
New Hampshire Avenue, Silver Spring, MD 20993
Christine Tesseromatis, Department of Pharmacology,
Medical School, Athens University, M Assias 75,
11527, Athens, Greece
Leslie M Tompkins, Department of Pharmaceutical
Sci-ences, School of Pharmacy, University of Maryland, 20
Penn Street, Baltimore, MD 21201
James A Uchizono, Department of Pharmaceutics and
Medicinal Chemistry, Thomas J Long School of
Phar-macy and Health Sciences, University of the Pacific,
Stockton, CA 95211
Hongbing Wang, Department of Pharmaceutical Sciences,
School of Pharmacy, University of Maryland, 20 Penn
Street, Baltimore, MD 21201
Jeffrey Wang, Department of Pharmaceutical Sciences,
College of Pharmacy, Western University of Health
Sciences, 309 E Second Street, Pomona, CA 91766
∗Deceased.
Stephen Wang, Drug Metabolism and Pharmacokinetics,
Merck Research Laboratories, 2015 Galloping HillRoad, Kenilworth, NJ 07033
Current Affiliation: DMPK/NCDS, Millennium: TheTakeda Oncology Company, 35 Landsdowne Street,Cambridge, MA 02139
Lori B Karpes, Department Industrial and Physical
Phar-macy, College of PharPhar-macy, Nursing and Health ences, Purdue University, West Lafayette, IN 47907
Sci-Lauren Wiser, Department of Pharmaceutics and
Medici-nal Chemistry, Thomas J Long School of Pharmacy andHealth Sciences, University of the Pacific, Stockton, CA95211
Cindy Q Xia, Drug Metabolism and
Pharmacokinet-ics, Millennium Pharmaceuticals, Inc., 35 LandsdowneStreet, Cambridge, MA 02139
Wen Xie, Center for Pharmacogenetics and Department of
Pharmaceutical Sciences, University of Pittsburgh, 633Salk Hall, 3501 Terrace Street, Pittsburgh, PA 15216
Jaime A Y´a ˜nez, Department of Drug Metabolism and
Pharmacokinetics (DMPK), Alcon Laboratories, Inc.,
6201 S Freeway, Fort Worth, TX 76134
Bin Zhang, Center for Pharmacogenetics and Department
of Pharmaceutical Sciences, University of Pittsburgh,Pittsburgh, PA 15216
Lei Zhang, Offices of Clinical Pharmacology, Center
for Drug Evaluation and Research, Food and DrugAdministration, Building 51, Room 3106 10903 NewHampshire Avenue, Silver Spring, MD 20993
Current affiliation: Frontage Laboratories, Inc., Exton,
PA 19341
Li Zhang, School of Pharmacy, Faculty of Medicine, The
Chinese University of Hong Kong, Hong Kong
Yuanchao (Derek) Zhang, Offices of Clinical
Pharmacol-ogy, Center for Drug Evaluation and Research, Food andDrug Administration, Building 51, Room 3106 10903New Hampshire Avenue, Silver Spring, MD 20993Current affiliation: Frontage Laboratories, Inc., Exton,PA
Deliang Zhou, Manufacturing Science and Technology,
Global Pharmaceutical Operations, Abbott Laboratories,North Chicago, IL
Wei Zhu, Department of Pharmaceutical Sciences and
Clinical Supplies, Merck and Co., Inc., 770 SumneytownPike, P.O Box 4, WP 75B-210, West Point, PA 19486
Zhong Zuo, School of Pharmacy, Faculty of Medicine,
The Chinese University of Hong Kong, Hong Kong
Trang 15Portal vein Blood
Bile
Bypass hepatocytes
Trang 16NAT FMO
2D6 2E1 3A4(+5)
2B6
2C19 2C9(+8)
2D6 2E1 3A4(+5)
2B6
2C19 2C9(+8)
2D6 2E1 3A4(+5)
2B6
2C19 2C9(+8)
2D6 2E1 3A4(+5)
2B6
2C19 2C9(+8)
2D6 2E1 3A4(+5) (a)
Figure 8.1 (a) Contribution of individual human enzyme systems to metabolism of marketed
drugs; (b) contribution of individual P450s in metabolism of drugs UGT indicates
uridinedinu-cleotide phosphate (UDP) glucuronosyl transferase; FMO, flavin-containing monooxygenase; NAT,
N-acetyltransferase; MAO, monoamine oxidase; P450, cytochrome P450 Source: Adapted from
2%
(b) (a)
CYP1A2 CYP2B6 CYP2C CYP2D6 CYP2E1 CYP3A4 other
researchers identify new substrate drugs Source: Adapted from Wang and Tompkins (2008).
Trang 17SULT1A1 FMO5
OATP2 Carboxylesterase CYP7A
XRE
CYP1A CYP1B UGT1A1 UGT1A3 UGT1A6 BCRP
Figure 10.6 Target genes of PXR, CAR, and AhR (a) PXR and CAR are shown dimerized totheir common partner, RXR and sitting response elements XREM (xenobiotic response enhancermodule) and PBREM (phenobarbital response enhancer module), respectively Overlapping targetgenes are boxed in the center with CAR-specific targets shown above and PXR-specific targetsshown below (b) AhR is shown bound to partner, ARNT and activating its target genes afterbinding to the XRE (xenobiotic response element; often called the dioxin response element).UGT1A1 represents a common target gene of all three receptors
OAT4
PepT1/2 OCTN2 OAT1
OAT3
MATE-1/2
MRP2/4 PgP
Trang 18BCR BCRP P-gp MRP
MCT
MRP OCT
POT Intracellular accumulation
POT?
Nucleus
E
TAP-1 TAP-2
R
Passive Transcellular Diffusion
Abluminal
Figure 18.1 A representative depiction of a number of transporters expressed in a human intestinal
cell illustrating the complexity of the system Source: Modified from Carl et al (2007).
Trang 19r
Efflux pump
Prodrug not recognized
S I D E Biotransformation of prodrug to drug
Enzymes or chemicals
Drug
Prodrug
Transporter r
Transporter ATP
ATP
Efflux pump
Drug not recognized
Figure 22.1 Role of efflux and influx transporters in intestinal absorption
Trang 20Paracellular transport
Carrier mediated transport
e l l
c y
Tight cell junction
Carrier mediated efflux transporter
y t o p l a s
ATP
Active
transport
Transporter Facilitator protein Efflux pump
Figure 22.3 Intestinal drug transport mechanisms
(a)
(b)
7
1 2 6
7 27
CYP2C8 CYP2C9 CYP2C19 CYP2D6 CYP3A Non-CYP Phase I
41
40 19 3 16 47
13
39
1A1 1A3 1A4 1A6 1A8 (extrahepatic) 1A9
1A10 (extrahepatic) 2B7
2B15 Other
Figure 26.2 (a) Distribution of CYP and non-CYP phase I enzyme pathways for 65 oral drugs(NMEs approved between 2003 and 2008) (3) (b) Distribution of UGT enzyme pathways reported
for 103 drugs from the literature (Kiang et al., 2005) and Drugs@FDA Most of them are expressed
in the liver except for UGT1A8 and UGT 1A10
Trang 21Figure 28.4 Simplified structure of P-gp structure and function.
CDF-Mrp-2 Substrate
Ca 2+ buffer pretreated for 10 min Ca 2+ buffer pretreated for 10 min
+MK571 (Ca2+buffer pretreated for 10 min) +MK571 (Ca2+ buffer pretreated for 10 min)
MK571: a Mrp-2 inhibitor
BC BC
Figure 30.6 Fluorescence and phase-contrast micrographs of hepatocytes treated with CDFDA
in the presence and absence of MK571 These results demonstrate that depletion of Ca2 + opensthe tight junction and enables compounds to be released from bile canaliculi (BC) MK571, whichdid not disrupt BC, blocked the excretion of CDF into BC via its inhibitory effect on Mrp2
Trang 22(wild type)
m PXR knockout
h PXR
transgene
(loss-of-function)
(humanized function) (gain-of-function)
Figure 32.2 Strategies to create the loss-of-function knockout, gain-of-function transgenic, and
the combined “humanized” function models Source: Adapted from Gong et al (2005), with the
permission of the publisher
Trang 231.1 INTRODUCTION
Oral bioavailability of a drug is a measure of the rate and
extent of the drug reaching the systemic circulation and is
a key parameter that affects its efficacy and adverse effects
Therefore, study of oral bioavailability has received
consid-erable attention in scientific arena Unfortunately, we are
unable to predict bioavailability as a priori to this date,
although we have made significant progress in
understand-ing various components of this complex puzzle, includunderstand-ing
solubility (e.g., aqueous solubility), partition coefficients
(e.g., octanol/water), absorption (e.g., permeability across
the Caco-2 cell membrane), metabolism (e.g.,
microsome-mediated phase I metabolism), and excretion (e.g., efflux
via p-glycoprotein) However, understanding a few of these
components would not allow us to accurately predict a
drug candidate’s bioavailability in humans Therefore, oral
bioavailability remains to be a highly experimental
param-eter that eludes prediction from modern computational
or experimental approaches, although some preliminary
Oral Bioavailability: Basic Principles, Advanced Concepts, and Applications, First Edition Edited by Ming Hu and Xiaoling Li.
© 2011 John Wiley & Sons, Inc Published 2011 by John Wiley & Sons, Inc.
progress has been made in recent years Continued progress
to develop a better and more thorough understanding ofphysicochemical and biochemical profiling of drug or drug-like molecules would be needed to alleviate the problemsassociated with bioavailability, and some progress has beenmade in the last decade (Ho and Chien, 2009) Poor oralbioavailability is also one of the leading causes of fail-ures in clinical trials This is because compounds withlow bioavailability would have a highly variable expo-sure between individuals If a compound has an averagebioavailability of 5%, it would easily vary in the range of0.5–10%, a 20-fold difference This difference makes theselection of an appropriate dose particularly difficult sincetoo little may yield no impact and too much could result intoxicity, which is not acceptable for most drugs that desirechronic administration
The reasons why oral bioavailability is such a lenge for development of drugs or drug-like substances(e.g., nutraceuticals) are several-fold: first, many physic-ochemical and biological factors contribute to the bioavail-ability of a compound; second, many scientific disciplinesare involved but few, if any, scientists are good at morethan one specific area; third, reliable scaling from ani-mal models to humans is often absent; and fourth, oralbioavailability is often seriously affected by diet andpolypharmacy, neither of which can be adequately con-trolled in a standard clinical trial, considering the diversity
chal-of the population— the elderly and seriously ill patients
In addition, we are normally able to gain access only tolimited body fluids such as blood and urine, and fluidssurrounding the target tissues/cells are often not accessi-ble This limitation makes bioavailability, a measure of theextent and rate of absorption and the elimination processes,
1
Trang 242 BARRIERS TO ORAL BIOAVAILABILITY—AN OVERVIEW
bile and kidney, so other elimination route (e.g., exhalation) is not shown (See insert for color representation of the figure.)
really representing only systemic blood exposure to drugs
(Fig 1.1) Therefore, it is not surprising that bioavailability
would sometimes not satisfactorily correlate with efficacy
Oral bioavailability remains a major challenge to
the development of nutraceuticals and naturally derived
chemopreventive agents For example, many scientists are
interested in developing plant-derived polyphenols into
chemopreventive agents Polyphenols are derived from
plants and consumed in the form of fruits, vegetables,
spices, and herbs In different regions of the world, a
large percentage of dietary polyphenols are consumed
in the form of flavonoids from various sources of food
intake, although cultural and dietary habit dictates which
forms of polyphenols are consumed (Fletcher, 2003; Slavin,
2003; Aggarwal et al., 2007) On the other hand, a large
percentage of population do not take sufficient quantities
of fruits and vegetables for a variety of reasons (Adhami
and Mukhtar, 2006) Therefore, scientists are interested
in developing a pill that will mimic the effects of
ingesting fruits and vegetables Yet, today their effort
has not produced a single polyphenolic chemopreventive
agent; the unsuccessful attempt may be attributed to the
poor bioavailability of polyphenols (usually <5%) Poor
bioavailability makes the evaluation of a chemopreventive
agent a particular challenge, since the clinical trials for
chemopreventive agents often involve a large population
for a prolonged period and extremely high costs
When all of the above-mentioned challenges are takeninto consideration in the product development of drugs orchemopreventive agents, it is obvious that developing anappropriate oral dosage form for drug candidate or can-didate of chemopreventive agent is not a trivial or straightforward task Although pharmaceutical scientists have greatdifficulty in predicting and enhancing bioavailability, thereward is also immense as the vast majority of top rev-enue and prescription leaders are orally administered drugs.Therefore, we devote this chapter to briefly introduce each
of the factors that influence bioavailability and guide thereaders to the appropriate chapters in this book where theycan obtain in-depth contents of each related topic
As an oral dosage form enters the oral cavity and thenthe gastrointestinal (GI) tract, several barriers must beovercome before it can reach the systemic circulation andthe therapeutic target On its way to the therapeutic target,
a drug in a given dosage form will need to first overcomethe preabsorption barrier formed by the hostile acidic andenzymatic environment in the stomach and intestine Thenthe drug would encounter the primary barrier formed bythe biological membrane, that is, the wall of the GI tract.Once a drug successfully passes the intestinal epitheliumbarrier, the drug will need to overcome another barrierconsisting of transporters and enzymes, which utilize theefflux mechanism to pump the drug back to the intestineand degrade the drug via the first-pass effect There are
Trang 25many factors that will affect a drug molecule’s ability
to overcome these barriers to reach and remain in the
systemic circulation These factors include the inherent
physicochemical properties of the drug molecules,
biologi-cal characteristics of the GI tract, pathophysiologibiologi-cal state,
drug–drug or drug–food interactions, etc
1.1.1 Physicochemical Factors
Various physicochemical factors will affect the oral
bioavailability of a drug The importance of
physicochemi-cal properties of a drug molecule in drug absorption or
per-meation was illustrated by Lipinski’s “rule of 5” (Lipinski
et al., 2001) Because of the importance of
physicochemi-cal properties, a thorough characterization of drug substance
would provide fundamental information for drug discovery,
as well as for formulation and dosage form development
The characterization of key physicochemical properties of
drug substances is described in Chapter 2 One of the key
physicochemical properties that play a crucial role in the
drug absorption/permeation is solubility Solubility defines
the maximum concentration of a drug available for
absorp-tion or permeaabsorp-tion, while another important
physicochem-ical property, dissolution rate, controls the rate of the drug
available for absorption or permeation Factors that affect
solubility and dissolution rate surely will also influence the
bioavailability of the drug Variation of pH in the GI tract
causes drugs to behave differently in terms of solubility and
dissolution rate along the GI tract For an acidic drug, a low
solubility and slow dissolution rate in the stomach, where
pH is low, can be expected, while for a basic drug, poor
sol-ubility owing to precipitation in the intestinal fluids, where
pH is high, would happen An understanding of the basic
concept of solubility and dissolution rate forms a solid
foun-dation for comprehending bioavailability Physicochemical
factors also dictate the permeability of drug molecules
Solubility and permeability of a drug are such important
factors for drug absorption or bioavailability The combined
effect of these two factors would determine the
developa-bility and bioavailadevelopa-bility of a compound to a certain extent
Chapters 3 and 4 discuss the two important factors related
to drug absorption, namely, solubility and dissolution rate
Chapter 6 provides the fundamentals for drug permeation
or absorption Chapter 7 correlates the physicochemical
parameters in vitro and in vivo.
1.1.2 Biological Factors
Oral delivery is a preferred route for the administration
of small molecule drugs, because the intestine has a very
large surface area, in excess of 200 m2, which is the
size of a tennis court Since oral absorption is limited by
the drugs with molecular weight <600 Da and effective
absorption window in the GI tract, permeability of drug
through intestinal membrane, physiology of GI tract, andmetabolism of drugs in absorption and transport havebecome important factors with respect to bioavailability
GI tract is not always a hospitable place for drugabsorption Enzymes are secreted in the GI tract at arate of about 45 g per day in adult humans Althoughthe primary functions of these enzymes are to digestnutrients such as protein, carbohydrates, and nucleotides,their presence is one of the primary reasons why proteinand genetic materials (for gene therapy) cannot be deliveredorally, unless special formulation approaches are used
In addition to surviving in the hostile environment, adrug needs to overcome the barriers posted by theintestinal epithelium Intestinal epithelium is a complextissue with advanced cellular structures and metabolicfunctionality The presence of cellular junctions, especiallytight junction, severely impedes the passage of molecules
with molecular weight >200 Da via the paracellular route.
Therefore, the vast majority of the drug molecules mustuse the transcellular route Transcellular route is affected
by a myriad of interrelated but sometimes competingbiological factors Although it was always believed thatlipophilic molecules have an easy access to the transcellularroute, the presence of various efflux transporters thatpreferentially bind with lipophilic molecules could seriouslylimit the absorption of lipophilic molecules In addition,
if a molecule is too lipophilic (e.g., log P > 5), it may
be retained in the cellular membrane Because intestinalepithelial cells have a functional existence of only three
to four days (near or at the tip of the intestinal villus),molecules that bind too tightly will be eventually lostwhen the epithelial cells slough off Hydrophilic drug
molecules with molecular weight >200 Da cannot penetrate
the intestinal epithelium by passive diffusion; they musthave special structural motifs that make them attractive forthe nutrient transporters such as amino acid transporters(Chapter 17), the small peptide transporter 1 (or PepT1)(Chapter 18), organic ion transporters (Chapter 19), andnucleobases transporters Assuming drug molecules getinto the epithelial cells, there are intestinal first-passmetabolisms capable of further degrading their chance
to reach the systemic circulation These metabolisms areprimary phase II metabolism although CYP3A4 is thought
to be decently active in the enterocytes In Chapters 5, 6,
8, and 10, the barriers to oral bioavailability have beendescribed in greater details, with emphasis on GI biology(Chapters 5 and 16), drug absorption (Chapter 6) andmetabolism pathways (Chapter 8), and drug excretion bythe enterocytes (Chapter 9)
The last major barrier to oral bioavailability, perhapsthe most well-known one, is the first-pass metabolism inthe liver Since all drugs absorbed via the GI tract (exceptthe last few centimeters of the rectum) have to enter theportal vein and encounter hepatocytes (each of which can
Trang 264 BARRIERS TO ORAL BIOAVAILABILITY—AN OVERVIEW
be called metabolic superstar ), escaping liver metabolism
is the last step in the oral absorption process
In addition to these important factors, protein binding,
which affects drug distribution and free drug available for
metabolism, has also featured in this book (Chapter 11)
Lastly, another general factor that affects the systemic
exposure, elimination via urine, is discussed in Chapter 12
Taken together, substantial information is provided on the
pharmacokinetic behaviors of drugs following oral
admin-istration (Chapter 13), many of which can be explained
using the information learned from previous chapters
1.1.3 Diet and Food Effects
Development of drugs is becoming more global and
multi-dimensional The day where a standard diet is appropriate
for clinical trials across the globe is probably over
Tradi-tionally, diet and food effects have focused on the protein
content, caloric intake, and fat amounts, and few if any have
carefully examined the effects of other more exotic dietary
components such as spices More recently, consumers are
taking ever large quantities of dietary supplements with
increasing frequency and variety Although we are unable
to completely address how these changes in the diet will
impact drug bioavailability, various attempts have been
made Chapter 14 has shed some light on this topic
1.1.4 Drug Interactions
Drug interactions remain a serious concern for the
develop-ment of new drugs On the basis of the target patient
popula-tion, certain types of drug interactions are not acceptable to
the manufacturer, FDA (Food and Drug Administration of
the United States of America), or both Traditionally, drug
interactions are classified into pharmacodynamic
interac-tions and pharmacokinetic interacinterac-tions and this book mainly
deals with the latter in Chapter 15, since it is a book focused
on oral bioavailability
Classical pharmacokinetic drug interactions typically
involve phase I metabolic enzymes, and clinical examples
of this type of interactions are well documented in the
literature From a pharmacokinetic point of view, drug
interaction may cause a rise or a fall in body exposure of
drug, that is, change in Cmax(maximal drug concentration)
and/or AUC (area under the curve) values From a
mechanistic point of view, a rise in exposure is typically
related to inhibition of enzyme activities or down regulation
of relevant metabolic enzymes, whereas a fall in exposure
is typically related to activation of dormant enzymes or
induction of relevant metabolic enzymes
More recently, FDA is contemplating the inclusion of
efflux transporters into the drug interaction universe, and
provisional guidance has been issued This could further
complicate the drug development process and increase
the complexity and cost of development The reasons areseveral-fold First, many drugs undergo efflux and phase
I metabolism simultaneously and therefore it is difficult
to sort out the precise mechanisms of drug interactions.Second, there are few demonstrated clinical cases whereinteractions with efflux transporters have been confirmed
as the sole source of drug interactions Third, metabolicenzymes may develop significant interplay with the effluxtransporters such that it would be necessary to interactwith both components of the disposition in order to displayclinically significant effects Many of these are discussed
in Chapter 26
1.1.5 Formulation Factors
Based on the physicochemical and biological factors thataffect the bioavailability, we can use different strategies toovercome the barriers for bioavailability (Chaubal, 2004).One can design a dosage form that can avoid the harshenvironment in the stomach or optimally utilize the absorp-tion window For example, an enteric-coated dosage formwill not dissolve until it reaches the intestine while a gas-troretentive drug delivery system can prolong the residenttime of dosage forms in the GI tract Oral dosage forms can
be coated with rate-limiting membranes that can controlthe rate of drug release from the dosage form Increasedsolubility and dissolution rate are effective ways to improvebioavailability One can create an effective dissolution ratethat supplies the proper amount of soluble drug for absorp-tion Nanoparticles are increasingly becoming an importantpart of modern drug dosage form design as incorporation ofnanoparticles can often alleviate challenges associated withpoor solubility Varieties of pharmaceutical technologiesand drug delivery approaches have been used to improvethe physicochemical properties of the drugs Approachesfor various dosage form design and solubility/dissolutionenhancement can be found in Chapters 20 and 21.Enhancement of solubility and dissolution rate allowsformulation scientists to manipulate the factors related todrug substances for improving bioavailability To improvebioavailability, one can also modulate the permeability ofdrug across the intestinal epithelium Chapters 22 and 23represent some of the attempts in this direction
1.2 SCIENTIFIC DISCIPLINES INVOLVED
It was once thought that the intestinal tract is a very modating organ for drug absorption because this organ isbuilt to absorb nutrients If drugs are good for us, shouldthe intestinal tract be there to do what benefits us? It wasnot until 1990s that the myth—if medicinal chemists can
accom-develop active compounds in vitro, formulation scientists can make a finished product to deliver them in vivo —was
found to be untrue Development of two classes of
Trang 27compounds, renin inhibitors and HIV protease inhibitors,
convinced drug development scientists and senior
manage-ment in pharmaceutical companies that oral bioavailability
matters because the intestine is not just an absorption organ
Scientists with various training and education
back-grounds are involved in the development of orally
administered drugs Aside from classical biologists and
medicinal chemists that are involved in the drug
discov-ery phase, more preclinical ADME (absorption,
distribu-tion, metabolism, and excretion) works are now integrated
into the drug discovery area Once a candidate is selected,
additional ADME work plus toxicology will be needed
to further advance the candidate into clinical trials Then,
physician researchers, nurse practitioners, biostatisticians,
marketing professionals, and pharmaceutical economists
become involved in the clinical studies At the same time, a
different group of scientists, many with engineering
back-ground, are making decisions on the manufacturing and
processing parameters Therefore, it is not entirely
unex-pected that scientists in different groups do not always
have the proper background to fully understand each other
One of the purposes of this book is to provide an
easy-to-understand section for the scientists in different areas to
understand ADME and their terminologies
One of the important goals of this book is to give a
prac-tical guide to the use of several state-of-the-art technologies
and methodologies to readers who may or may not be
famil-iar with these techniques in order to gain a basic
understand-ing and knowledge for practical approaches and/or methods
The readers can dive into the contents ranging from
advanced reviews on various important efflux transporters
that affect drug absorption and excretion (Chapter 24),
to the coupling between efflux transporters and enzymes
(Chapter 25), to computational methods and approaches to
predict bioavailability (Chapter 33) For the technology and
methodology, the readers can find a detailed description on
the Caco-2 cell culture model (Chapter 27), MDCK (Madin
Darby canine kidney and other related cell culture models
(Chapter 28), intestinal perfusion (Chapter 29), liver
perfu-sion (Chapter 30), primary hepatocytes (Chapter 30), in vivo
pharmacokinetics (Chapter 31), and methods to determine
regulation of enzymes and transporters (Chapter 32)
Rapid advances in the human genomics and proteomics
promise to better predict factors determining human
responses to drugs Although the price of sequencing the
whole human genome remains out of the practical range at
present, rapid advances in this area are expected to make
the practice an economic reality in the not so distant future
Recent passage of a law by the Congress of the United
States of America to ban discrimination based on genetic
information should provide the legal framework to protect
an individual’s right to utilize his/her genetic information
for better health care This law and progress in the
economics of human genome sequencing will mean that,
in the near future, we could develop criteria that will dosepatients according to his/her genetic makeup—a radicalprogress in the field of individualized pharmacy We willall welcome the day that geneticists become active partic-ipants in the drug development process, instead of limitingtheir participation only in the drug discovery process
Oral bioavailability remains a big challenge for smallmolecules, and an even bigger challenge for macromolec-ular drugs such as protein Despite decades of effort, there
is no product for oral insulin This book devotes itself tothe study of various biological and physicochemical prin-ciples and methodologies that can be used to understandthe oral bioavailability problems and to devise strategiesthat can be used to overcome these problems Although
we still cannot predict bioavailability as a priori at this
time, it is getting closer to the moment when we would beable to do so for the small molecular drugs Efforts under-taken by various drug delivery companies are on the brink
of achieving oral delivery of active insulin Therefore, thescience of oral bioavailability is closer than ever in the his-tory of drug development to become an enabler of drugdevelopment, instead of an obstacle to drug development.Together with the advent of individualized genomic infor-mation, we are heading to a day when each of the patientscould receive drug according to his/her conditions We areall very hopeful that this day is within our grasp in the nearfuture
REFERENCES
Adhami VM and Mukhtar H (2006) Polyphenols from green tea
and pomegranate for prevention of prostate cancer Free Radic
Res 40:1095–1104.
Aggarwal BB, Sundaram C, Malani N, and Ichikawa H (2007)
Curcumin: the Indian solid gold Adv Exp Med Biol 595:1–75.
Chaubal MV (2004) Application of drug delivery technologies in
lead candidate selection and optimization Drug Discov Today
9:603–609.
Fletcher RJ (2003) Food sources of phyto-oestrogens and their
precursors in Europe Br J Nutr 89(Suppl 1):S39–43.
Ho RJ and Chien JY (2009) Drug delivery trends in clinical trialsand translational medicine: Updated analysis of ClinicalTri-
als.gov database J Pharm Sci 98:1928–1934.
Lipinski CA, Lombardo F, Dominy BW, and Feeney PJ (2001)Experimental and computational approaches to estimate sol-ubility and permeability in drug discovery and development
settings Adv Drug Deliv Rev 46:3–26.
Slavin J (2003) Why whole grains are protective: biological
mechanisms Proc Nutr Soc 62:129–134.
Trang 282.2.1 Ultraviolet/visible (UV/vis) diffuse
2.3.3 X-ray powder diffractometry (XRPD) 12
2.3.4 Thermal methods of analysis 13
2.3.5 Thermogravimetric analysis (TGA) 13
2.3.6 Differential thermal analysis (DTA) 13
2.3.7 Differential scanning calorimetry (DSC) 13
Oral Bioavailability: Basic Principles, Advanced Concepts, and Applications, First Edition Edited by Ming Hu and Xiaoling Li.
© 2011 John Wiley & Sons, Inc Published 2011 by John Wiley & Sons, Inc.
2.1 INTRODUCTION
Solid dosage forms, such as tablets and capsules, are a mon means of administration of pharmaceutically active
com-ingredient (API) in humans (Gennaro, 1985; Brittain et al.,
1991; Brittain, 1995) They are manufactured by ing a number of powdered solids together, most com-monly, blending or mixing of multiple components, milling
process-or size reduction, granulation which may be done eitherusing a granulating fluid or in the dry state (roller com-paction), compression into tablets and coating (Lachman
et al., 1986) All these processes may be influenced by the
physical properties of the solids, and, thus, their tance is being increasingly recognized A number of testshave been included in the United States Pharmacopeia(USP) to characterize these physical properties of pow-dered solids (The United States Pharmacopeia and NationalFormulary, 2002) It is important to have a comprehensiveunderstanding of the physical characteristics earlier duringproduct development to prevent future problems such aswas observed with ritonavir In this case, a new, more ther-modynamically stable, less soluble polymorph was being
impor-formed during bulk drug manufacturing after the product
was launched in the market As a consequence, stability
and bioavailability of the product were at risk (Bauer et al.,
2001)
The API and excipients used in formulations can exist
in various physical phases that can impact processability,
7
Trang 29stability, and performance of the formulation, which have
been briefly summarized below
2.1.1 Crystalline and Amorphous Phases
Crystalline materials are characterized by a regular,
well-defined, and long-range (>20 A) periodicity in the˚
arrangement of the constituent atoms, ions, or molecules
They exhibit sharp melting points and characteristic
X-ray powder patterns Amorphous materials lack long-range
order in their molecular arrangement They do not have
melting points and their X-ray patterns show a broad halo
The most common route of obtaining amorphous solids is
by rapid cooling of a melt below its melting point where
the structural characteristics of the liquid are maintained
but the viscosity is much higher This is considered as
a “supercooled liquid.” On further cooling, a
character-istic temperature known as the glass transition
tempera-ture (Tg )is observed below which the solid is “kinetically
frozen” into an unstable glassy state with properties
dif-ferent from both the supercooled liquid and crystalline
form Amorphous solids possess higher free energy than
their crystalline counterparts due to which they have higher
apparent solubilities, dissolution rates, and enhanced
chem-ical reactivities (Hancock and Zografi, 1997) For example,
the amorphous form of sulfapyridine was found to have a
higher apparent solubility and dissolution rate than its
crys-talline counterpart (Gouda et al., 1977) When the same
dose was administered to dogs, therapeutically adequate
concentrations were obtained with amorphous novobiocin
while the crystalline form was not absorbed at all This
difference in bioavailability was attributed to the
differ-ences in apparent solubility and dissolution rates between
the amorphous and crystalline phases (Mullins and Macek,
1960)
2.1.2 Polymorphic Forms
Polymorphism is the ability of a compound to crystallize
as two or more phases having different arrangements
and/or molecular conformations in the crystal lattice
(Brittain, 1999) Polymorphs of a given compound are
chemically identical but, in general, different in structure
and properties including dissolution rates, melting point,
density, hardness, and crystal shape The choice of the
polymorphic form may determine the physical and chemical
stability, compressibility, and bioavailability of the drug
substance (Haleblian et al., 1971) A compound can exist
in a number of polymorphic forms but only one form
is thermodynamically stable at a given temperature and
pressure, while the others are metastable The metastable
polymorphs have higher free energies, apparent solubilities,
and dissolution rates than their stable counterparts The
larger the free energy difference between the stable
and metastable polymorphs, the higher is the expecteddifference in solubility (Brittain, 1999) For example, whendifferent polymorphs of tolbutamide (forms I, II, III, andIV) were administered to dogs, forms II and IV showedhigher bioavailabilities than forms I and III (Kimura
et al., 1999) While it is generally preferred to formulate
the most thermodynamically stable form, under somecircumstances, it may be desirable to use the metastableforms in formulations because of their higher dissolutionrates However, owing to higher reactivity of metastablephases, their physical, as well as chemical, stability needs
to be carefully monitored during processing and storage
2.1.3 Solvates
Solvates are adducts or molecular complexes containingsolvent molecules within the crystal structure in either sto-ichiometric or nonstoichiometric proportions If the incor-porated solvent molecule is water, the solvate is referred
to as a hydrate (Vippagunta et al., 2001) The
incorpora-tion of the solvent molecule in the crystal lattice results
in differences in physical and pharmaceutical properties.Differences in solubility of the hydrated and anhydrousphases may result in a difference in bioavailability Whenanhydrous ampicillin and ampicillin trihydrate were admin-istered to dogs, the peak serum levels following adminis-tration of the anhydrate were higher and occurred earlier
(Poole et al., 1968) Similar results were observed after
their oral administration to humans (Ali and Farouk, 1981).Similarly, differences in the solubility of the anhydrousand the dihydrate phases of carbamazepine resulted indifferences in bioavailability when administered to dogs.The bioavailability of carbamazepine dihydrate was lowerthan that of the corresponding anhydrate forms (Kobayashi
et al., 2000).
The discussion above indicates that it is important tocharacterize materials to select and control the desirable
form in formulations Brittain et al (1991) have defined
a systematic approach for physical characterization ofpharmaceutical solids where the material properties wereclassified as molecular, particulate, and bulk properties Theobjective of this chapter is to summarize the techniquesused for physicochemical characterization of solids based
on this classification This chapter provides only a briefoutline and the reader is encouraged to go through thereferences for a deeper understanding of the subject
2.2 MOLECULAR LEVEL PROPERTIES
These are defined as those characteristics that could bemeasured at a molecular level They include spectroscopictechniques and are based on properties such as molecularinteractions and molecular bond energies These studies
Trang 30MOLECULAR LEVEL PROPERTIES 9
can be performed at a very early stage with the advantage
of using minimum amount of material and providing
information regarding polymorphic form, solvate phase,
and crystallinity
2.2.1 Ultraviolet/Visible (UV/vis) Diffuse Reflectance
Spectroscopy
Although UV/vis techniques are widely used for the
anal-ysis of solutions, it can be adapted for the
character-ization of solids This is performed by using diffuse
reflectance techniques instead of in the transmission mode
It is associated with the fraction of radiation that
pene-trates into the bulk and then emerges (Brittain, 1995) The
instrumentation consists of a light source, a
monochro-mator, an integrating sphere, and a detector The
instru-ment and sample preparation may be optimized to
mini-mize undesirable specular reflectance (surface reflectance
of the incident beam) Several diffuse reflectance
theo-ries have been proposed but the Kubelka –Munk theory
is the most generally accepted Diffuse reflectance can be
k and s = the molar absorption and the scattering
coefficients, respectively, and
R∞= the reflectivity of an infinitely thick sample
The equation is valid in weakly absorbing systems
with a small particle size (∼1 μm) without
signif-icant contribution from specular reflectance (Kubelka,
1948)
UV/vis diffuse reflectance spectroscopy has been used
to evaluate solid–solid interactions in formulations The
effect of formulation composition and processing variables
on the microenvironment in solid dosage forms was
eval-uated using indicator probes providing a measure of the
physicochemical nature of the formulation in the solid state
(Govindarajan et al., 2006a) The technique has also been
used to correlate pH of the solution before freeze-drying and
chemical reactivity of the freeze-dried material
(Govindara-jan et al., 2006b), and evaluation of the Maillard reaction
between a primary amine and lactose (Wu et al., 1970).
2.2.2 Vibrational Spectroscopy
Infrared (IR) and Raman spectroscopies are complementary
techniques widely used for the characterization of
phar-maceutical solids The IR region in the electromagnetic
spectrum can be divided into three regions; the near-IR
(4000–14,000 cm−1), mid-IR (400– 4000 cm−1), and
far-IR (100– 400 cm−1) with the near- and mid-IR generally
being used for analysis When a sample is irradiated,absorption of IR energy results in transitions betweenmolecular vibrational and rotational energy levels (either ofsingle pairs of atoms or groups of atoms) The molecularvibrations depend on the structure of the analyte and thuscan be used for the identification of a molecular identity
IR and Raman spectroscopies complement each other byevaluating various functional groups While polar groupssuch as C O and NH are likely to be IR active, bondssuch as C C and SS are more likely to be Raman active(Brittain, 1995)
Spectrometers usually consist of an electromagneticsource, a sample chamber, and a detector For IR anal-ysis, sample preparation can be carried out using differ-ent methods including (i) alkali halide pellet, where theanalyte is pulverized with either KBr or KCl and com-pressed into discs; (ii) mull preparation, where the analyte
is mixed with ∼1 mg of mineral oil; (iii) use of a neatpowdered sample in diffuse reflectance infrared Fouriertransform spectroscopic technique (DRIFTS), it is nonin-vasive but is particle size dependent; (iv) attenuated totalreflectance (ATR), where the sample is placed in contactwith an IR transmitting crystal with a high refractive indexthrough which the IR beam is directed and then penetrates
a few micrometers into the sample; and (v) tic spectroscopy—modulated IR radiation is selectivelyabsorbed by the sample Similar to DRIFTS, no samplepreparation is required for Raman spectroscopy where anal-ysis can be carried out on small samples of the neat mate-rial Both IR and Raman spectroscopes can be combinedwith an optical microscope enabling analysis of a few crys-tals (Bugay, 2001)
photoacous-Since IR and Raman techniques are based on themolecular structure of materials, they can be used forthe analysis of polymorphs and solvates Five forms
of tranilast (three polymorphs and two solvates) werecharacterized by using IR and Raman spectroscopes Whileform II was determined to be a conformationally distinctivepolymorph, form III had a different packing with weakerintermolecular hydrogen bonds Owing to its ease andrapidity, IR spectroscopy was used to determine polymorph
contamination during process development (Vogt et al.,
2005) Some authors have reported the use of thesetechniques for the analysis of materials with differentdegrees of crystallinity (Okumura and Otsuka, 2005)and solid dispersions (Konno and Taylor, 2006) Ramanspectroscopy was used to detect amorphous indomethacin
at a level of 2% w/w using a calibration curve ofindomethacin with various degrees of crystallinity Agrowing application of NIR spectroscopy is in the field
of process analytical technologies (PATs) such as real-timemonitoring of blending processes (El-Hagrasy and Drennen,2005)
Trang 312.2.3 Solid-State Nuclear Magnetic Resonance
(SSNMR)
Nuclear magnetic resonance (NMR) spectroscopy has been
extensively used to analyze molecules in the solution phase
The use of NMR for the study of solids is now being widely
used for the characterization of pharmaceutical solids
There are several interactions that a nucleus with a magnetic
moment undergoes when placed in a magnetic field Some
of these include Zeeman interactions (interaction between
magnetic moment of the nucleus and the external magnetic
field), dipole –dipole interaction (magnetic coupling of two
nuclei through space), chemical shift (magnetic shielding by
surrounding electrons), and spin–spin couplings (indirect
coupling between two spins) A thorough discussion of
all these interactions is outside the scope of this chapter;
therefore, references are provided (Brittain, 1995; Tishmack
et al., 2003).
There are, however, some differences in the analysis of
solutions and solids The strong dipolar interactions and
chemical shift anisotropy (CSA) in solids lead to broad
peaks in the spectrum These are not observed in solutions
because of the fast random motions of the samples CSA
occurs in solids since the orientation of the molecules
is fixed with respect to the magnetic field These issues
can be overcome by probes that can be subjected to high
decoupling power and by rapidly spinning the sample
Magic angle spinning (MAS) can also be used wherein
the line width can be minimized by spinning the sample
at an angle of 54.7◦with the magnetic field (Brittain, 1995;
Tishmack et al., 2003).
Solid-state nuclear magnetic resonance (SSNMR) has
been used for the qualitative and quantitative
character-ization of solids that can exist as polymorphs, solvates,
or in the amorphous state MAS NMR was used for the
evaluation of several polymorphs and hydrates of
olanza-pine (Fig 2.1) (Reutzel-Edens et al., 2003) The spectra
were characterized by highly resolved, sharp resonances
The unique chemical shifts reflected the presence of the
carbon nuclei in different polymorphic forms and states of
hydration Presence of form II in form III as an impurity
was observed using X-ray diffraction (XRD) and this was
confirmed by using SSNMR (Reutzel-Edens et al., 2003).
In a review by Shah et al (2006), several examples are
discussed where SSNMR was used for the evaluation of
amorphous forms of drugs
2.2.4 Solubility
Solubility is defined as the concentration of the dissolved
solid in a solvent medium in a saturated solution in
equilibrium with the solid at a defined temperature and
pressure (Martin et al., 1983) The time dependence of this
solubilization process is often measured, which is referred
Chemical Society Copyright© 2003
to as dissolution testing (Grant and Higuchi, 1990) (see
Chapter 4 for details) Various dissolution testing methods
of dosage forms have been reported in the USP Some ofthe factors affecting solubility include the physical form
of the solid, the nature and composition of the solvent,temperature, and pressure (The United States Pharmacopeiaand National Formulary, 2002) Solubility is the topic
of discussion in Chapter 3, and therefore has not beendiscussed in detail here
Trang 32PARTICULATE LEVEL PROPERTIES 11
However, it is important to recognize the influence of the
physicochemical characteristics on the solubilization and
dissolution rate of API from a dosage form At a given
temperature, the metastable phases such as the amorphous
form or metastable polymorphic form will theoretically
have a higher dissolution rate than the corresponding
stable form attributed to the free energy differences (Grant
and Higuchi, 1990) The anhydrate form of theophylline
has an apparent solubility twice that of its hydrate in
aqueous media due to its higher thermodynamic activity
However, at equilibrium, its solubility was determined
to be the same as that of the hydrate due to its
conversion to the hydrate form (Shefter and Higuchi, 1963)
Phadnis and Suryanarayanan (1997) have discussed the
impact of incorporating the metastable form of anhydrous
theophylline in tablets Owing to recrystallization of the
stable form on storage, the dissolution of these tablets failed
the USP dissolution test In a literature review by Pudipeddi
and Serajuddin (2005), solubility ratios of polymorphs
and anhydrates/hydrates of a number of APIs have been
reported Typically, it was determined that the solubility
ratio of polymorphs was less than 2 Some examples, where
the solubility differences have resulted in differences in
bioavailability, have been discussed in Section 2.1
2.3 PARTICULATE LEVEL PROPERTIES
These are properties that are associated with individual solid
particles that can be characterized by evaluating a small
amount of powder
2.3.1 Particle Morphology
A crystal is bounded by a number of planar faces and
the arrangement of these faces is termed habit (Brittain,
1995) Particulates can be classified into the following
based on their different crystal habits (The United States
Pharmacopeia and National Formulary, 2002):
1 acicular—needle-like particles, width and thickness
are similar;
2 columnar—rod-like particles, larger and thicker than
acicular particles;
3 flake —thin flat particles;
4 plate —flat particles with similar length and width,
thicker than flakes;
5 lath—thin blade-like particles;
6 equant—particles of similar length, width, and
thick-ness
These different particle morphologies can impact the
flow of materials, dissolution characteristics, and
compres-sion properties An evaluation of various habits of celecoxib
showed that the lath-shaped crystals had better flow and
compressibility characteristics (Banga et al., 2007)
Swami-nathan and Kildsig (2000) demonstrated that the surfaceroughness of particle carriers had a significant effect on thestability of powder mixtures Mixtures containing highlytextured surfaces segregated to a lesser extent than thosewith a smoother texture probably because of a higher num-ber of sites of adhesion In the case of dipyridamol [crystalmodification of dipyridamol using different solvents andcrystallization conditions (Adhiyaman and Basu, 2006)],crystals with different morphology were produced by crys-tallizing from various solvents These crystals exhibiteddifferent dissolution rates Similar observations have beenreported for phenytoin where the dissolution rate increasedbecause of inclusion of dopants improving wettability by ahabit-related increase in the polar crystal face areas (Chow
et al., 1995).
Particle morphology can be evaluated by optical andelectron microscopic techniques Widely used opticalmicroscopes include the light and polarizing microscopes.The light microscope simply consists of a magnifying lenswith a stage to mount the sample A polarizing microscope
is a light microscope with two polarizers The technique
is based on the effect the analyte has on the transmittedlight For further details, the reader is encouraged to review
the work of Kuhnert-Brandst¨atter (1971), McCrone et al.
(1978), and Brittain (1995)
Scanning electron microscopy is widely used for uating particle morphology of pharmaceuticals It has anelectron gun generating an electron beam, a column withlenses for beam focusing, the sample chamber, and detector.When the electron beam interacts with an inner electron ofthe sample it is ejected with a discrete amount of energy
eval-This is referred to as the secondary electron These
sec-ondary electrons escaping from the surface of the sampleare attracted to the detector, accelerated, and visible light isproduced The emitted light is detected by a photomultipliertube (Brittain, 1995)
2.3.2 Particle Size Distribution
Particle size distribution of the API and the excipientsplays an important role during the process of mixing orblending The magnitude of the gravitational and inertialforces is largely determined by the particle size of thecomponents being blended Most powders having meanparticle size below 100 μm are not free flowing due
to high interparticulate forces (Lachman et al., 1986).
Owing to these forces, blending is more uniform when thecomponents have similar particle sizes Some techniquesthat can be used for the determination of particle sizedistribution include sieve analysis, microscopy, and lightscattering techniques The USP has described the methodfor powders where at least 80% of the particles are larger
Trang 33than 75 μm Sieves are stacked on top of one another in
ascending degrees of coarseness, and the test powder is
placed on the top sieve The nest of sieves is subjected
to a standardized period of agitation, and the weight of
the material retained on each sieve is determined, which
provides the weight percentage of powder in each sieve
size range The size parameter involved in determining
particle size distribution by analytical sieving is the length
of the side of the minimum square aperture through which
the particle passes (The United States Pharmacopeia and
National Formulary, 2002)
Particle size of the powders can influence several
properties of formulations such as content uniformity
where it was observed that the narrower the particle
size distribution and smaller the particle size, the better
is the content uniformity (Zhang and Johnson, 1997)
Rohrs et al (2006) developed a method for determining
the particle size distribution width and the dose required
to conform with the content uniformity requirements of
the USP Literature reports also discuss the influence of
particle size on powder flow (Podczeck and Newton, 1999;
Kachrimanis et al., 2005) and granulation and tabletting
properties (Sun and Grant, 2001; Herting and Kleinebudde,
2007) Generally, powders with smaller particle sizes yield
tablets with a higher tensile strength In the study by
Sun and Grant (2001), different size fractions of l-lysine
monohydrochloride dihydrate were compressed Crystals
with smaller particle size resulted in compacts with higher
tensile strength at a given pressure attributable to the larger
number of contact points with the smaller sized particles
Dissolution rate is inversely proportional to the particle size,
that is, the smaller the particle size of the API, the higher
is the dissolution rate (Florence and Salole, 1976; Elamin
et al., 1994).
In a recent publication, Shekunov et al (2006) have
reviewed the various methods of measuring particle size
distributions Laser diffraction is being widely used in the
pharmaceutical industry due to its robustness, short
mea-surement time, high precision, and flexibility Generally,
such instruments use a standard He –Ne laser light source
and the diffracted light is detected by a position-sensitive
detector The measurement is strongly influenced by the
shape of the particles Some other techniques used for the
measurement of particle size distribution include dynamic
light scattering (used for emulsions and colloids), the
coul-ter councoul-ter (used for nonagglomerated systems), and the
Anderson Cascade Impactor (for inhalations)
2.3.3 X-ray Powder Diffractometry (XRPD)
X-ray powder diffractometry (XRPD) is an excellent
tech-nique for the characterization of crystalline phases Every
crystalline solid phase has a unique X-ray pattern which can
form the basis for its identification In a powder mixture,
each crystalline phase produces its pattern independently ofthe other constituents in the mixture Thus, XRPD enablesnot only the identification of one API in the presence ofexcipients, but also the simultaneous identification of morethan one API in formulations (Brittain, 1995)
An X-ray diffractometer exposes the sample to
electro-magnetic radiation lying between the ultraviolet and γ -rays
in the electromagnetic spectrum When the X-rays are dent on the crystalline solids, diffraction occurs, that is,the X-rays are scattered in all directions A peak in theX-ray pattern is observed when these scattered beams are
inci-in phase and reinci-inforce each other, and this is definci-ined byBragg’s law Thus, when a monochromatic beam of wave-
length λ is incident on a sample at an angle θ diffraction
occurs, if
nλ = 2d sin θ
d is the distance between the planes in the crystal (angstrom
units) and n is the order of reflection The USP also
provides an introduction to X-ray diffractometry (Brittain,1995; The United States Pharmacopeia and NationalFormulary, 2002)
XRPD can be used to identify different crystalline formssuch as hydrates and polymorphs Various crystalline solidforms of fluprednisolone have been characterized by usingXRPD In this study, XRPD was used for the identification
of (i) anhydrous polymorphic forms; (ii) solvates; and (iii)anhydrous form existing in both crystalline and amorphous
states (Haleblian et al., 1971) Phadnis and Suryanarayanan
(1997) have evaluated the phase transformations occurringduring processing and storage of anhydrous theophyllinetablets and its implication of tablet dissolution using XRPD
40
Figure 2.2 XRD patterns of (a) stable anhydrous theophylline I,(b) stable theophylline monohydrate II, and (c) metastable anhy-
drous theophylline I* (Phadnis et al., 1997) Source: Reprinted
with permission from Wiley-Liss, Inc, a subsidiary of John Wileyand Sons, Inc Copyright© 1997
Trang 34PARTICULATE LEVEL PROPERTIES 13
Quantitative analysis of various components in a
formulation can be carried out using XRPD When
appro-priate absorption corrections are made, the peak intensities
of each component are proportional to the weight fraction
of that component in the mixture This method was used
for determining the amounts of anhydrous carbamazepine
and carbamazepine dihydrate in a mixture (Suryanarayanan,
1989) Low levels of contamination of α-carbamazepine
(<5% w/w) in β-carbamazepine could also be detected and
quantified (Phadnis et al., 1997) XRPD can also be used
for the quantification of crystalline material in the
pres-ence of the amorphous form as was reported by Surana and
Suryanarayanan (2000) Crystalline sucrose was detected
and quantified in a mixture with amorphous sucrose up to
levels of 1–5% w/w of the crystalline form
XRPD can also be carried out under a controlled
envi-ronment enabling the monitoring of phase transformations
as they occur The temperature (variable temperature PXRD
(powder X-ray diffraction)) and the water vapor pressure
of the chamber can be changed to evaluate phase
trans-formations Solute crystallization in a ternary system
dur-ing all stages of freeze-drydur-ing was monitored by X-ray
while simulating the process in situ in the chamber of
the diffractometer (Pyne et al., 2003) The influence of
both temperature and water vapor pressure on the
dehy-dration of carbamazepine dihydrate was evaluated by using
variable temperature XRPD While X-ray amorphous
car-bamazepine was formed at water vapor pressures of ≤5.1
Torr, crystalline anhydrous γ -carbamazepine was formed
at≥12 Torr Thus, XRPD in these controlled environments
helped elucidate the anhydrous phases obtained under
dif-ferent dehydration conditions (Han et al., 1998).
2.3.4 Thermal Methods of Analysis
Thermal analysis is a group of techniques in which
a property of the sample is measured against time or
temperature while the temperature of the sample, in a
specified environment, is changed This temperature change
may be linear, or the sample property may be measured
when the temperature is held constant These techniquesare very useful in preformulation studies where the APIproperties such as polymorphism, state of hydration, degree
of crystallinity, purity, and degradation characteristics can
be measured (Brittain, 1995)
2.3.5 Thermogravimetric Analysis (TGA)
It is a measure of the thermally induced weight loss
of a material as a function of applied temperature It
is a useful method for the quantitative analysis of totalvolatile content of a solid Therefore, it can be used tocharacterize desolvation and degradation processes In arecent study, the dehydration of calcium benzoate hydrateswas evaluated using thermogravimetric analysis (TGA).The weight loss observed during TGA corresponded tothe stoichiometric water content of the hydrates (Fig 2.3;Terakita and Byrn, 2006) The influence of grinding onthe loss of acetonitrile from quinapril hydrochloride wascharacterized using TGA (Han and Suryanarayanan, 1997;
Guo et al., 2000) It is a complementary technique to
differential scanning calorimetry (DSC) and differentialthermal analysis (DTA)
2.3.6 Differential Thermal Analysis (DTA)
The difference in temperature between a sample and
a reference as a function of temperature is monitored.Therefore, if the sample undergoes an endothermic reaction,its temperature will lag behind that of the reference.However, in the case of an exothermic reaction itstemperature will exceed that of the reference DTA has beenmost commonly used to determine temperatures at whichthermal events occur In the above study by Terakita andByrn (2006), DTA was used as a complementary techniquefor the characterization of dehydration (Fig 2.3)
2.3.7 Differential Scanning Calorimetry (DSC)
DSC is widely used for the characterization of ceuticals, and the USP also has a general chapter on this
pharma-30
60 90 120 150 Temperature (°C)
0 2 4 6 8 10 12
–20
–15 –10 –5 0 5 10 15 20
Figure 2.3 TGA/DTA scans of (a) calcium benzoate trihydrate and (b) monohydrate using an openpan at 5◦C/min (Terakita and Byrn, 2006) Source: Reprinted with permission from Wiley-Liss,Inc., a subsidiary of John Wiley and Sons, Inc Copyright© 2006
Trang 35Exotherm corresponding to degradation Glass transition with enthalpic recovery
Figure 2.4 DSC curves (heating rate of 10◦C) of TGME samples milled for 30, 60, and 300 s
(Shalaev et al., 2002) Source: Reprinted with permission from Wiley-Liss, Inc., a subsidiary of
John Wiley and Sons, Inc Copyright© 2002
technique (The United States Pharmacopeia and National
Formulary, 2002) This method of analysis is similar to
DTA There are two types of measurement—power
com-pensated and heat flux While in power-comcom-pensated DSC,
the sample and the reference are kept at the same
temper-ature (heat flow required to maintain the same tempertemper-ature
is monitored), in heat flux DSC the temperature
differen-tial between the sample and the reference is monitored
DSC can be used to monitor thermal events, including
melting point, crystallization, desolvation, and glass
tran-sitions The impact of milling on the thermal properties
of tetraglycine methyl ester (TGME) was characterized by
using DSC (Fig 2.4) Change in the melting endotherm
due to milling was observed indicating creation of a
dis-order An endotherm associated with enthalpic recovery at
the glass transition temperature was also observed (Shalaev
et al., 2002) Thus, the study exhibits the utility of the
tech-nique for characterizing several phase transformations, that
is, melting, glass transition, and degradation For further
understanding of the technique, the reader is referred to the
review by Clas et al (1993).
2.3.8 Modulated DSC
In this technique, a controlled temperature modulation is
overlaid on the conventional linear heating or cooling
rate to produce a continuously changing nonlinear sample
temperature This helps in separating out overlapping
thermal events such as enthalpic recovery and glass
transition (Royall et al., 1998).
2.3.9 Pressure DSC
The sample is subjected to different pressures, and thus
used to separate overlapping endotherms, for example,
dehydration and vaporization of water The use of this
technique to characterize the dehydration of carbamazepinedihydrate and ampicillin trihydrate has been demonstrated
by Han and Suryanarayanan (1997, 1998), respectively
2.4 PROPERTIES ASSOCIATED WITH THE BULK LEVEL
These include all the bulk material properties includingparticle shape, size distribution, powder flow, and density.The science and technology of small particles is referred to
as micromeritics as proposed by DallaValle (1948) These
particle properties can strongly influence various processesduring manufacturing, and hence knowledge about theseproperties and control over them are important Severaltechniques are available for characterizing these properties
2.4.1 Surface Area
The surface area per unit weight or volume can influencedissolution, chemical reactivity, and bioavailability of theAPI In the case of excipients, flowability is a major factorthat is influenced by the surface area; for example, grades
of lactose and Avicel with larger particles, thus smallersurface area have better flow properties (DallaValle, 1948;
Brittain et al., 1991) The surface area can be calculated
from the particle size distribution described earlier Someother techniques available for its direct determination aredescribed below
Adsorption Method
For the determination of the surface area of an adsorbent,the volume of gas (in cubic centimeters) can beplotted against the pressure of the gas adsorbed pergram of adsorbent The BET equation can be used to
Trang 36PROPERTIES ASSOCIATED WITH THE BULK LEVEL 15
more accurately determine the volume of nitrogen gas
V m in cubic centimeters that 1 g of the powder can
adsorb when a monolayer is complete (Martin et al.,
1983)
Air Permeability Method
The resistance to the flow of air through a plug of
compacted powder is proportional to the surface
area of the powder Therefore, the permeability
across a given pressure drop in the plug is inversely
proportional to the specific surface area The plug
of powder can be regarded as a series of capillaries
and the internal surface depends on the particle
surface area The flow rate depends on the surface
area of the particles, the degree to which the
particles are compressed, and the irregularity of
the capillaries The Kozeny–Carman equation can
be used to determine the surface area using this
A= the cross-sectional area of the plug,
K = a constant,
ε= the porosity,
V = volume of air flowing through the
capillary of length l in t seconds, and
η = the viscosity of air (Martin et al., 1983).
2.4.2 Porosity
The porosity of a solid is defined as the ratio of the
void volume to the bulk volume (total sample volume)
and is frequently represented as a percentage Porosity
can influence the dissolution of both powders and tablets
For instance, an insoluble drug may dissolve more or less
rapidly in aqueous medium depending on their adsorption
of moisture or air (Martin et al., 1983) Properties such
as powder flow (Ohta et al., 2003) and dissolution rate of
tablets are also influenced by its pore structure and porosity
(Otsuka et al., 2007) Pore radii and volume measurements
can be carried out by using either gas adsorption or mercury
porosimetry (Martin et al., 1983) For mercury porosimetry,
the sample is placed in a holder with a tapered calibrated
stem The sample holder and stem are then filled with
mercury and pressure is applied to force the mercury into
the pores The amount of mercury that penetrates the sample
can be determined from the decrease in volume in the
calibrated stem, which is indicative of sample porosity
(Martin et al., 1983).
2.4.3 Density
Density is defined as the weight per unit volume.Pharmaceutical processes, including material transfer,blending, and flow (for example, through a hopper or afeeder), can be significantly influenced by the density of thecomponents Reports in the literature discuss the influence
of the difference in densities on segregation, and thus
con-tent uniformity issues during manufacturing (Rippie et al.,
1964; Venables and Wells, 2001)
Three types of densities can be defined:
is calculated from the weight of the liquid that thesample displaces In flotation, a few crystals of thepowder are suspended in the liquid whose density isclose to that of the material and the temperature isvaried till the crystals float It is at this temperature,that the density of the powder and the liquid arethe same and thus the density of the powder can
be determined All these techniques are discussed indetail by Duncan-Hewitt and Grant (1986)
Granule or Particle Density
It is the weight of the particles per unit volume asdetermined by mercury intrusion (considering poreslarger than 10 μm) It is determined by a methodsimilar to liquid displacement Mercury is used as theliquid since it can penetrate the intraparticle space but
not the internal pores of the particles (Martin et al.,
1983)
Bulk Density
The volume occupied by a powder is referred to as
the bulk volume Density determined from the bulk
volume and weight of the powder is referred to
Trang 37as bulk density The bulk density depends on the
size, shape, porosity, and cohesiveness of powders
For smaller particles (<10 μm), flow may be
restricted because the cohesive forces are equal to
the gravitational forces Flat particles such as needles
and flakes tend to pack loosely to give powders
with high porosity Poor flow may also arise due to
high moisture content and surface roughness (which
increases friction and cohesiveness) One of the
most widely used methods to determine bulk density
is to pour the powder into a graduated cylinder
and measure the bulk volume and the weight of
the powder Tap density is also often measured by
subjecting the powder in the graduated cylinder to a
number of measured taps, for example, 500 or 1000
Another variation of this measurement involves the
tapping of the cylinder for specified number of taps,
until a constant volume is obtained (Martin et al.,
1983)
These powder characteristics such as the density,
particle size distribution, and shape can significantly
influence the flow of powders or granules, as discussed
so far Pharmaceutical processability such as blending,
encapsulation, and compression depend on the flowability
of the granules Several methods can be used to measure
the flowability of powders
2.4.4 Angles of Repose
This is the angle formed between cone of a powder and
the horizontal plane Powders with lower angles of repose
exhibit better flow The rougher and more irregular the
surface of the particles, the higher will be the angle of
repose Particles with more spherical shape have a lower
angle of repose (Martin et al., 1983).
2.4.5 Compressibility or Carr’s Index
This can be calculated using the following equation:
v = the tapped volume (after being subjected to a
specified number of taps) and
v0 = the bulk volume (Lachman et al., 1986).
Generally, values lower than 15% indicate good flow,
whereas values above 25% indicate poor flowability
Several examples exist in literature that illustrate the
utility of Carr’s Index to evaluate the flow characteristics
(Podczeck and Newton, 1999; Nagel and Peck, 2003)
2.4.6 Water Sorption
Sorption of water by pharmaceutical solids can have
an impact on both their chemical and physical stability.Water sorption/desorption isotherms can be generatedeither gravimetrically or volumetrically For gravimetricmeasurements, a specific weight of the material is placed on
a balance in a chamber or environment whose temperatureand relative humidity are controlled When the solid doesnot sorb any more moisture (i.e., it has a constant weight),the relative humidity and/or the temperature of the samplemay be changed Thus, the moisture sorption at eachhumidity condition can be monitored This is referred
to as the continuous measurement of moisture sorption.
Figure 2.5 presents the continuous water vapor sorptionprofiles of amorphous trehalose prepared by differentmethods (freeze-dried, spray dried, melt quenched, and
dehydrated) (Surana et al., 2004) Alternatively, different
samples of the solid can be placed in chambers wherethe humidity is controlled by saturated salt solutions Thesamples are removed periodically and weighed, until thesample is at equilibrium (i.e., its weight does not change
between weighings) This is referred to as the discontinuous
method (Brittain, 1995).
Solids can come in contact with water due to contactwith hygroscopic excipients, either during processing (such
as wet granulation) or during storage (from moisture
in the head space) At low relative humidities, whilecrystalline solids absorb moisture on the surface, watervapor is absorbed into the bulk of amorphous material
As the humidity increases, multilayer sorption may occurand at a characteristic RH the solid may dissolve in thesorbed film As discussed earlier, the amorphous form of
0 0.0
2.0 4.0
Dehydrated I
Dehydrated II
Spray-dried 8.0
RH (%)
Figure 2.5 Water vapor sorption profiles of amorphous trehalose
prepared by different methods (Surana et al., 2004) Source:
Reprinted with permission from SpringerLink Copyright© 2004
Trang 38REFERENCES 17
solids tend to sorb more moisture than their crystalline
counterparts Certain pharmaceutical processes, including
milling, compaction, and spray drying, may induce a change
in the physical form of the API creating a local disorder
(Vadas et al., 1991) These high-energy spots may sorb
moisture to a greater extent than the crystalline state These
regions can undergo considerable change and affect the bulk
properties of the material
While this chapter provides a brief introduction to some
of the widely used techniques for the physicochemical
characterization of pharmaceuticals, references are provided
for a deeper understanding Several examples are provided
to understand the implication of these properties during
manufacturing It should be emphasized that an early
systematic characterization of material properties helps in
avoiding issues during formulation development
ACKNOWLEDGMENT
The author would like to thank Dr Raj Suryanarayanan,
Dr Sophie-Dorothee Clas, Dr Hubert Dumont, Dr Raghu
Cavatur, Ms Cecilia Madamba, and Ms Mary-Lynn Gaal
for their contributions to this chapter
REFERENCES
Adhiyaman R and Basu SK (2006) Crystal modification of
dipyri-damol using different solvents and crystallization conditions
Int J Pharm 321:27–34.
Ali AA and Farouk A (1981) Comparative studies on the
bioavailability of ampicillin anhydrate and trihydrate Int J
Pharm 9:239–243.
Banga S, Chawla G, Varandani D, Mehta BR, and Bansal AK
(2007) Modification of the crystal habit of celecoxib for
improved processability J Pharm Pharmacol 59:29–39.
Bauer J, Spanton S, Henry R, Quick J, Dziki W, Porter W,
and Morris J (2001) Ritonavir: an extraordinary example of
conformational polymorphism Pharm Res 18:859–866.
Brittain HG (1995) Physical Characterization of Pharmaceutical
Solids Marcel Dekker, Inc., New York.
Brittain HG (1999) Polymorphism in Pharmaceutical Solids.
Marcel Dekker Inc., New York
Brittain HG, Bogdanowich SJ, Bugay DE, DeVincentis J, Lewen
G, and Newman A (1991) Physical characterization of
pharmaceutical solids Pharm Res 8:963–973.
Bugay D (2001) Characterization of the solid-state: spectroscopic
techniques Adv Drug Del Rev 48:43–65.
Chow AHL, Hsia CK, Gordon JD, Young JWM, and
Vargha-Butler EI (1995) Assessment of wettability and its relationship
to the intrinsic dissolution rate of doped phenytoin crystals Int
J Pharm 126:21–28.
Clas S-D, Dalton C, and Hancock B (1993) Differential scanning
calorimetry: applications in drug development Pharm Sci Tech
Today 2:311–320.
DallaValle JM (1948) Micromeritics Pitman Publishing
Com-pany, New York
Duncan-Hewitt WC and Grant DJW (1986) True density andthermal expansivity of pharmaceutical solids: comparison
of methods and assessment of crystallinity Int J Pharm
characterization of powder mixing kinetics J Pharm Sci
Gennaro AR (1985) Remington’s Pharmaceutical Sciences Mack
Publishing Company, Pennsylvania
Gouda WM, Ebian AR, Moustafa MA, and Khalil SA (1977)
Sul-phapyridine crystal forms Drug Dev Ind Pharm 3:273–290.
Govindarajan R, Zinchuk A, Hancock B, Shalaev E, and narayanan R (2006a) Ionization states in the microenvironment
Surya-of solid dosage forms: effect Surya-of formulation variables and
pro-cessing Pharm Res 23:2454–2468.
Govindarajan R, Chatterjee K, Gatlin L, Suryanarayanan R, andShalaev E (2006b) Impact of freeze-drying on ionization ofsulfonephthalein probe molecules in trehalose-citrate systems
ride J Pharm Sci 89:128–143.
Haleblian JK, Koda RT, and Biles JA (1971) Isolation andcharacterization of some solid phases of fluprednisolone
J Pharm Sci 60:1485–1488.
Han J, Gupte S, and Suryanarayanan R (1998) Applications
of pressure differential scanning calorimetry in the study of
pharmaceutical hydrates II Ampicillin trihydrate Int J Pharm
carbamazepine dihydrate Pharm Dev Tech 3:587–596.
Hancock BC and Zografi G (1997) Characteristics and significance
of the amorphous state in pharmaceutical systems J Pharm Sci
86:1–12.
Trang 39Herting MG, Kleinebudde P (2007) Roll compaction/dry
granula-tion: effect of raw material particle size on granule and tablet
properties Int J Pharm 338:110–118.
Kachrimanis K, Petrides M, and Malamataris S (2005) Flow rate
of some pharmaceutical diluents through die-orifices relevant
to mini-tableting Int J Pharm 303:72–80.
Kimura K, Hirayama F, and Uekama K (1999) Characterization
of tolbutamide polymorphs (Burger’s forms II and IV) and
polymorphic transition behavior J Pharm Sci 88:385–391.
Kobayashi Y, Ito S, Itai S, and Yamamoto K (2000)
Physicochemi-cal properties and bioavailability of carbamazepine polymorphs
and dihydrate Int J Pharm 193:137–146.
Konno H and Taylor LS (2006) Influence of different polymers
on the crystallization tendency of molecularly dispersed
amorphous felodipine J Pharm Sci 95:2692–2705.
Kubelka P (1948) New contributions to the optics of intensely
light scattering materials J Opt Sci Am 38:448–457.
Kuhnert-Brandst¨atter (1971) Thermomicroscopy in the Analysis of
Pharmaceuticals Pergamon Press, Oxford.
Lachman L, Lieberman HA, and Kanig JL (1986) The Theory and
Practice of Industrial Pharmacy Lea & Febiger, Philadelphia,
PA
Martin A, Swarbick J, and Cammarata A (1983) Physical
Pharmacy Lea and Febiger, Philadelphia, PA.
McCrone WC, McCrone LB, and Delly JB (1978) Polarized Light
Microscopy Ann Arbor Science Publishers, Ann Arbor.
Mullins JD and Macek TJ (1960) Some pharmaceutical properties
of novobiocin J Am Pharm Ass 49:245–248.
Nagel KM and Peck GE (2003) Investigating the effects
of excipients on the powder flow characteristics of
theo-phylline anhydrous powder formulations Drug Dev Ind Pharm
29:277–287.
Ohta KM, Fuji M, and Chikazawa M (2003) Effect of geometric
structure of flow promoting agents on the flow properties of
pharmaceutical powder mixture Pharm Res 20:804–809.
Okumura T and Otsuka M (2005) Evaluation of the
microcrys-tallinity of a drug substance, indomethacin, in a pharmaceutical
model tablet by chemometric FT-Raman spectroscopy Pharm
Res 22:1350–1357.
Otsuka M, Tanabe H, Osaki K, Otsuka K, and Ozaki Y
(2007) Chemoinformetrical evaluation of dissolution property
of indomethacin tablets by near-infrared spectroscopy J Pharm
Sci 96:788–801.
Phadnis NV, Cavatur RK, and Suryanarayanan R (1997)
Iden-tification of drugs in pharmaceutical dosage forms by X-ray
powder diffractometry J Pharm Bio Anal 15:929–943.
Phadnis NV and Suryanarayanan R (1997) Polymorphism in
anhydrous theophylline -implications on the dissolution rate
of theophylline tablets J Pharm Sci 86:1256–1263.
Podczeck F and Newton JM (1999) Powder filling into hard gelatin
capsules on a tamp filling machine Int J Pharm 185:237–254.
Poole JW, Owen G, Silverio J, Freyhof JN, and Roseman SB
(1968) Physicochemical factors influencing the absorption of
the anhydrous and trihydrate forms of ampicillin Current Ther
Res 10:292–303.
Pudipeddi M and Serajuddin ATM (2005) Trends in solubility of
polymorphs J Pharm Sci 94:929–939.
Pyne A, Chatterjee K, and Suryanarayanan R (2003) Solute
crystallization in mannitol-glycine systems-implications on
protein stabilization in freeze-dried formulations J Pharm Sci
92:2272–2283.
Reutzel-Edens SM, Bush JK, Magee PA, Stephenson GA, andByrn SR (2003) Anhydrates and hydrates of olanzapine: crys-tallization: solid-state characterization, and structural relation-
ships Cryst Growth Design 3:897–907.
Rippie EG, Olsen JL, and Faiman MD (1964) Segregationkinetics of particulate solid systems II Particle density: size
interactions and wall effects J Pharm Sci 53:1360–1363.
Rohrs BR, Amidon GE, Meury RH, Secreast PJ, King HM, andSkoug CJ (2006) Particle size limits to meet USP content
uniformity criteria for tablets and capsules J Pharm Sci
95:1049–1059.
Royall PG, Craig DQM, and Doherty C (1998) Characterization
of the glass transition of an amorphous drug using modulated
DSC Pharm Res 15:1117–1121.
Shah B, Kakumanu VK, and Bansal AK (2006) Analyticaltechniques for quantification of amorphous/crystalline phases
in pharmaceutical solids J Pharm Sci 95:1641–1665.
Shalaev E, Shalaeva M, and Zografi G (2002) The effect ofdisorder on the chemical reactivity of an organic solid,tetraglycine methyl ester: change of the reaction mechanism
methods and applications Pharm Res 24:203–227.
Sun C and Grant DJW (2001) Effects of initial particle size on thetableting properties of l-lysine monohydrochloride dihydrate
powder Int J Pharm 215:221–228.
Surana R, Pyne A, and Suryanarayanan R (2004) Effect ofpreparation method on physical properties of amorphous
trehalose Pharm Res 21:1167–1176.
Surana R and Suryanarayanan R (2000) Quantitation of tallinity in substantially amorphous pharmaceuticals and study
crys-of crystallization kinetics by X-ray powder diffractometry
Powder Diffraction 15:2–6.
Suryanarayanan R (1989) Determination of the relative amounts
of anhydrous carbamazepine (C15H12N2O) and carbamzepinedihydrate (C15H12N2.2H2O) in a mixture by powder X-ray
diffractometry Pharm Res 6:1017–1024.
Swaminathan V and Kildsig DO (2000) The effect of particlemorphology on the physical stability of pharmaceutical powdermixtures: the effect of surface roughness of the carrier
on the stability of ordered mixtures Drug Dev Ind Pharm
26:365–373.
Terakita A and Byrn SR (2006) Structure and physical stability
of hydrates and thermotropic mesophase of calcium benzoate
J Pharm Sci 95:1162–1172.
Tishmack PA, Bugay DE, and Byrn SR (2003) Solid—statenuclear magnetic resonance spectroscopy-pharmaceutical
applications J Pharm Sci 92:441–474.
The United States Pharmacopeia and National Formulary (2002)United States Pharmacoepial Convention Inc., Rockville, MD.Vadas EB, Toma P, and Zografi G (1991) Solid-state phasetransitions initiated by water vapor sorption of crystalline
Trang 40Vippagunta SR, Brittain HG, and Grant DJW (2001) Crystalline
solids Adv Drug Del Rev 48:3–26.
Vogt FG, Cohen DE, Bowman JD, Spoors GP, Zuber GE,
Trescher GA, Dell’orco PC, Katrincic LM, Debrosse CW, and
Haltiwanger RC (2005) Structural analysis of polymorphism
and solvation in tranilast J Pharm Sci 94:651–665.
Wu W-H, Chin T-F, and Lach JL (1970) Interaction of
isoniazid with magnesium oxide and lactose J Pharm Sci
59:1234–1242.
Zhang Y and Johnson KC (1997) Effect of drug particle size on
content uniformity of low-dose solid dosage forms Int J Pharm
154:179–183.