You are on page 1of 41

Concepts and Models for Drug

Permeability Studies Cell and Tissue


based In Vitro Culture Models 1st
Edition Bruno Sarmento
Visit to download the full and correct content document:
https://textbookfull.com/product/concepts-and-models-for-drug-permeability-studies-c
ell-and-tissue-based-in-vitro-culture-models-1st-edition-bruno-sarmento/
More products digital (pdf, epub, mobi) instant
download maybe you interests ...

Stem Cell Derived Models in Toxicology 1st Edition Mike


Clements

https://textbookfull.com/product/stem-cell-derived-models-in-
toxicology-1st-edition-mike-clements/

Digital Business Models: Concepts, Models, and the


Alphabet Case Study Bernd W. Wirtz

https://textbookfull.com/product/digital-business-models-
concepts-models-and-the-alphabet-case-study-bernd-w-wirtz/

Molecular Evolutionary Models in Drug Discovery 1st


Edition Juan Bueno

https://textbookfull.com/product/molecular-evolutionary-models-
in-drug-discovery-1st-edition-juan-bueno/

In vitro bioassay techniques for anticancer drug


discovery and development 1st Edition Chandrashekhar H.

https://textbookfull.com/product/in-vitro-bioassay-techniques-
for-anticancer-drug-discovery-and-development-1st-edition-
chandrashekhar-h/
Cell Based Assays Using iPSCs for Drug Development and
Testing Carl-Fredrik Mandenius

https://textbookfull.com/product/cell-based-assays-using-ipscs-
for-drug-development-and-testing-carl-fredrik-mandenius/

Freight transport and distribution : concepts and


optimisation models Tolga Bektas

https://textbookfull.com/product/freight-transport-and-
distribution-concepts-and-optimisation-models-tolga-bektas/

Models for Sustainable Framework in Luxury Fashion


Luxury and Models 1st Edition Subramanian Senthilkannan
Muthu (Eds.)

https://textbookfull.com/product/models-for-sustainable-
framework-in-luxury-fashion-luxury-and-models-1st-edition-
subramanian-senthilkannan-muthu-eds/

Marketing Renewable Energy Concepts Business Models and


Cases 1st Edition Carsten Herbes

https://textbookfull.com/product/marketing-renewable-energy-
concepts-business-models-and-cases-1st-edition-carsten-herbes/

Patient Specific Induced Pluripotent Stem Cell Models


Generation and Characterization 1st Edition Andras Nagy

https://textbookfull.com/product/patient-specific-induced-
pluripotent-stem-cell-models-generation-and-characterization-1st-
edition-andras-nagy/
Concepts and Models for Drug Permeability
Studies
Related titles
Cell Culture Models of Biological Barriers: In Vitro Test Systems for Drug Absorption
and Delivery
(ISBN 978-0-415-27724-2)
Drug Absorption Studies: In Situ, In Vitro and In Silico Models
(ISBN 978-0-387-74900-6)
Woodhead Publishing Series in Biomedicine:
Number 79

Concepts and Models for


Drug Permeability Studies
Cell and Tissue Based In Vitro Culture
Models

Edited by

Bruno Sarmento

AMSTERDAM • BOSTON • CAMBRIDGE • HEIDELBERG


LONDON • NEW YORK • OXFORD • PARIS • SAN DIEGO
SAN FRANCISCO • SINGAPORE • SYDNEY • TOKYO
Woodhead Publishing is an imprint of Elsevier
Woodhead Publishing is an imprint of Elsevier
80 High Street, Sawston, Cambridge, CB22 3HJ, UK
225 Wyman Street, Waltham, MA 02451, USA
Langford Lane, Kidlington, OX5 1GB, UK

Copyright © 2016 Elsevier Ltd. All rights reserved.

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

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

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

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

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

ISBN: 978-0-08-100094-6 (print)


ISBN: 978-0-08-100114-1 (online)

British Library Cataloguing-in-Publication Data


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

Library of Congress Control Number: 2015939545

For information on all Woodhead Publishing publications


visit our website at http://store.elsevier.com/
Contents

List of contributors xi
List of figures xv
List of tables xxi

1 Introduction 1
Bruno Sarmento
1.1 Introduction 1
References 2

2 Importance and applications of cell- and tissue-based in vitro


models for drug permeability screening in early stages
of drug development 3
Miguel Ángel Cabrera Pérez, Marival Bermejo Sanz,
Victor Mangas Sanjuan, Marta González-Álvarez,
Isabel González Álvarez
2.1 Introduction 3
2.2 General considerations 4
2.3 Drug transport 4
2.4 Permeability–absorption models 13
2.5 Methods for permeability calculation 19
2.6 Standardization of protocols for in vitro methods 19
2.7 The “three Rs” principle 22
2.8 Biosecurity systems 24
References 24

3.1 Cell-based in vitro models for buccal permeability studies 31


Neha Shrestha, Francisca Araújo, Bruno Sarmento,
Jouni Hirvonen, Hélder A. Santos
3.1.1 Introduction 31
3.1.2 Physiology of the buccal mucosa 32
3.1.3 Different in vitro models 33
3.1.4 Conclusions 38
References 38

3.2 Cell-based in vitro models for gastric permeability studies 41


Tiago dos Santos, Bianca N. Lourenço, João Coentro, Pedro L. Granja
3.2.1 The stomach as a natural barrier to absorption 41
3.2.2 Gastric drug delivery 43
vi Contents

3.2.3 Cellularized models of gastric permeability 48


3.2.4 Conclusions 51
Acknowledgments 52
References 52

3.3 Cell-based in vitro models for intestinal permeability studies 57


Carla Pereira, Joana Costa, Bruno Sarmento, Francisca Araújo
3.3.1 Anatomy and physiology of human small intestine 57
3.3.2 Mechanisms of transport 59
3.3.3 Intestinal barriers 60
3.3.4 Intestinal in vitro models 61
3.3.5 Validation studies 73
3.3.6 Conclusions 75
References 75

3.4 Cell-based in vitro models for nasal permeability studies 83


Flávia Sousa, Pedro Castro
3.4.1 Introduction 83
3.4.2 Nasal primary cell culture models 84
3.4.3 Immortalized nasal cell lines 88
3.4.4 Nasal permeability studies 92
3.4.5 Conclusions 97
References 97

3.5 Cell-based in vitro models for pulmonary permeability studies 101


Fernanda Andrade, João Albuquerque, Ana Vanessa Nascimento
3.5.1 Introduction 101
3.5.2 Mechanisms involved in pulmonary absorption of drugs 102
3.5.3 Cell-based models of immortalized cells 104
3.5.4 Primary cell cultures 107
3.5.5 Conclusions 109
References 109

3.6 Cell-based in vitro models for vaginal permeability studies 115


Ingunn Tho, Natasa 
Skalko-Basnet
3.6.1 Introduction 115
3.6.2 Anatomy of the female genital tract and mucosa 116
3.6.3 Human primary cells 117
3.6.4 Immortalized human cells forming monolayers (bi-/tri-layers) 121
3.6.5 Commercially available three-dimensional culture
of nontransformed human vaginal-ectocervical epithelial cells 124
3.6.6 Concluding remarks 126
References 126
Contents vii

3.7 Cell-based in vitro models for ocular permeability studies 129


Teófilo Vasconcelos, Sara Baptista da Silva, Domingos Ferreira,
Manuela Pintado, Sara Marques
3.7.1 Introduction 129
3.7.2 Ocular anatomy 130
3.7.3 Ocular pharmacokinetics in the anterior segment 131
3.7.4 Ocular pharmacokinetics in the posterior segment 133
3.7.5 In vitro eye cellular models for drug permeability 134
3.7.6 Conclusions 147
References 148

3.8 Cell-based in vitro models for dermal permeability studies 155


Francisca Rodrigues, Maria Beatriz P.P. Oliveira
3.8.1 Introduction 155
3.8.2 Human skin and dermal permeability 156
3.8.3 Drug permeability in in vitro models 157
3.8.4 Reconstructed dermal equivalents 161
3.8.5 Reconstructed full-thickness models 163
3.8.6 Conclusions and future perspectives 165
References 165

3.9 Cell-based in vitro models for studying blood–brain


barrier (BBB) permeability 169
Maria João Gomes, Bárbara Mendes, Susana Martins,
Bruno Sarmento
3.9.1 Blood–brain barrier: structure, importance, and difficulties
to overcome 169
3.9.2 BBB in vitro models 171
3.9.3 Permeability of drugs: how to screen and study 183
3.9.4 Comparison of BBB models 184
Acknowledgments 185
References 185

4.1 Tissue-based in vitro and ex vivo models for buccal


permeability studies 189
Pedro Castro, Raquel Madureira, Bruno Sarmento,
Manuela Pintado
4.1.1 Introduction 189
4.1.2 Porcine buccal mucosa 190
4.1.3 Diffusion cells 194
4.1.4 Permeation assay using porcine buccal mucosa 196
4.1.5 Tissue integrity and viability assessment 197
4.1.6 Porcine esophageal mucosa 198
4.1.7 Conclusions and future prospects 199
References 199
viii Contents

4.2 Tissue-based in vitro and ex vivo models for intestinal


permeability studies 203
Rute Nunes, Cátia Silva, Luise Chaves
4.2.1 Introduction 203
4.2.2 Current tissue-based methodologies for intestinal
permeability studies 208
4.2.3 Animal versus human intestinal tissue 225
4.2.4 In vivo versus in vitro correlations 226
4.2.5 New trends in permeability studies using tissue-based
models 227
4.2.6 Conclusions 229
References 230

4.3 Tissue-based in vitro and ex vivo models for nasal


permeability studies 237
Alejandro Sosnik
4.3.1 Brief description of the structure of the nose 237
4.3.2 Nasal administration of drugs 238
4.3.3 Limitations of in vivo models 241
4.3.4 In vitro models of nasal permeability 242
4.3.5 Ex vivo models of nasal permeability 243
4.3.6 Conclusions 249
References 249

4.4 Tissue-based in vitro and ex vivo models for pulmonary


permeability studies 255
Ana Costa, Fernanda Andrade
4.4.1 Introduction 255
4.4.2 Lung physiology and tissue biology 256
4.4.3 Isolated perfused lung 260
4.4.4 Conclusions 267
References 267

4.5 Tissue-based in vitro and ex vivo models for vaginal


permeability studies 273
Alexandra Machado, José das Neves
4.5.1 Introduction 273
4.5.2 Vaginal permeability and absorption 274
4.5.3 In vitro 3D organotypic models 275
4.5.4 Ex vivo mucosal models 283
4.5.5 Conclusions 300
Acknowledgments 300
References 301
Contents ix

4.6 Tissue-based in vitro and ex vivo models for ocular


permeability studies 309
Christian Kölln, Stephan Reichl
4.6.1 Introduction 309
4.6.2 Requirements for a valid corneal cell culture model
for in vitro drug absorption studies 309
4.6.3 Methods to obtain corneal cells 310
4.6.4 Methods to verify cultivated cell layers in the construct 311
4.6.5 3D reconstructed cornea models 311
4.6.6 Discussions 320
4.6.7 Conclusions 321
References 321

4.7 Tissue-based in vitro and ex vivo models for dermal


permeability studies 325
Isabel Almeida, Paulo Costa
4.7.1 Introduction 325
4.7.2 Structure and function of the skin 326
4.7.3 Mechanisms of skin absorption 327
4.7.4 Mathematical principles of skin absorption 328
4.7.5 Conducting in vitro dermal absorption tests 330
References 338

4.8 Tissue-based in vitro and ex vivo models


for blood–brain barrier permeability studies 343
Malgorzata Burek, Ellaine Salvador, Carola Y. Förster
4.8.1 Introduction 343
4.8.2 Structure and function of BBB 343
4.8.3 Cerebral microvessels and their characteristics 345
4.8.4 Methods for cell isolation and immortalization 346
4.8.5 Cell-based in vitro BBB models and their properties
necessary for drug permeability estimation 346
4.8.6 Immortalized endothelial cell lines 347
4.8.7 Static and dynamic models of BBB compared 349
4.8.8 Measurements of drug permeability 350
4.8.9 Conclusions and future developments 351
References 351

5 Correlation between cell- and tissue-based in vitro models


for drug permeability screening with in vivo situation:
modeling and functional extrapolation 357
Paulo Paixão, Luís Gouveia, José Augusto Guimarães Morais,
Nuno Silva
5.1 Introduction 357
5.2 Empirical correlations 357
x Contents

5.3 Physiologically based pharmacokinetic models 361


5.4 Conclusions 367
References 368

Index 373
List of contributors

Jo~
ao Albuquerque University of Porto, Porto, Portugal
Isabel Almeida University of Porto, Porto, Portugal
Fernanda Andrade Laboratory of Pharmaceutical Technology, Faculty of
Pharmacy, University of Porto, Porto, Portugal; Institute for Bioengineering of
Catalonia (IBEC), Barcelona, Spain
Francisca Araujo Division of Pharmaceutical Chemistry and Technology, Faculty
of Pharmacy, University of Helsinki, Helsinki, Finland; INEB—Instituto de
Engenharia Biomédica, Universidade do Porto, Porto, Portugal; ICBAS—Instituto de
Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
Marival Bermejo Sanz Universidad Miguel Hernandez, San Juan de Alicante,
Alicante, Espa~
na
Malgorzata Burek Department of Anaesthesia and Critical Care, University of
Wurzburg, Wurzburg, Germany

Miguel Angel Cabrera Pérez Universidad Central Marta Abreu de Las Villas,
Santa Clara, Villa Clara, Cuba; Universidad Miguel Hernandez, San Juan de
Alicante, Alicante, Espa~na
Pedro Castro CBQF—Centro de Biotecnologia e Química Fina—Laboratorio
Associado, Escola Superior de Biotecnologia, Universidade Catolica Portuguesa/
Porto, Porto, Portugal
Luise Chaves UCIBIO/REQUIMTE—Laboratory of Applied Chemistry,
University of Porto, Porto, Portugal
Jo~
ao Coentro INEB—Instituto de Engenharia Biomédica, Universidade do Porto,
Porto, Portugal; Faculdade de Engenharia da Universidade do Porto (FEUP), Porto,
Portugal; Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do
Porto, Porto, Portugal
Ana Costa Instituto de Engenharia Biomédica (INEB), University of Porto, Porto,
Portugal; CESPU, Instituto de Investigaç~ao e Formaç~ao Avançada em Ciências e
Tecnologias da Sa
ude, Gandra PRD, Portugal
Joana Costa INEB—Instituto de Engenharia Biomédica, Universidade do Porto,
Porto, Portugal; FEUP—Faculdade de Engenharia da Universidade do Porto, Porto,
Portugal
xii List of contributors

Paulo Costa University of Porto, Porto, Portugal


Sara Baptista da Silva Faculty of Pharmacy, University of Porto, Porto,
Portugal; CBQF—Centro de Biotecnologia e Química Fina—Laboratorio
Associado, Escola Superior de Biotecnologia, Universidade Catolica Portuguesa/
Porto, Porto, Portugal
José das Neves INEB—Instituto de Engenharia Biomédica, Universidade do Porto,
Porto, Portugal; Instituto de Investigaç~ao e Inovaç~ao em Saude, Universidade do
Porto, Porto, Portugal
Tiago dos Santos INEB—Instituto de Engenharia Biomédica, Universidade do
Porto, Porto, Portugal
Domingos Ferreira Faculty of Pharmacy, University of Porto, Porto, Portugal
Carola Y. F€
orster Department of Anaesthesia and Critical Care, University of
Wurzburg, Wurzburg, Germany
Isabel Gonz 
alez Alvarez Universidad Miguel Hernandez, San Juan de Alicante,
Alicante, Espa~
na
Marta Gonz 
alez-Alvarez Universidad Miguel Hernandez, San Juan de Alicante,
Alicante, Espa~
na
Luís Gouveia iMed.UL, Faculty of Pharmacy, University of Lisbon, Lisbon,
Portugal
Pedro L. Granja INEB—Instituto de Engenharia Biomédica, Universidade do
Porto, Porto, Portugal; Faculdade de Engenharia da Universidade do Porto (FEUP),
Porto, Portugal; Instituto de Ciências Biomédicas Abel Salazar (ICBAS),
Universidade do Porto, Porto, Portugal
Jouni Hirvonen Division of Pharmaceutical Chemistry and Technology, Faculty
of Pharmacy, University of Helsinki, Helsinki, Finland
Maria Jo~ ao Gomes INEB—Instituto de Engenharia Biomédica, Universisdade do
Porto, Porto, Portugal; ICBAS—Instituto de Ciências Biomédicas Abel Salazar,
Universidade do Porto, Porto, Portugal; Instituto de Investigaç~ao e Inovaç~ao em
Saude, Universidade do Porto, Porto, Portugal
Christian K€
olln Institut f€
ur Pharmazeutische Technologie, Technische Universit€at
Braunschweig, Braunschweig, Germany
Bianca N. Lourenço INEB—Instituto de Engenharia Biomédica, Universidade do
Porto, Porto, Portugal; Faculdade de Engenharia da Universidade do Porto (FEUP),
Porto, Portugal
Alexandra Machado INEB—Instituto de Engenharia Biomédica, Universidade do
Porto, Porto, Portugal; Instituto de Investigaç~ao e Inovaç~ao em Saude, Universidade
do Porto, Porto, Portugal
List of contributors xiii

Raquel Madureira CBQF—Centro de Biotecnologia e Química Fina—Laboratorio


Associado, Escola Superior de Biotecnologia, Universidade Catolica Portuguesa/
Porto, Porto, Portugal
Victor Mangas Sanjuan Universidad Miguel Hernandez, San Juan de Alicante,
Alicante, Espa~
na
Sara Marques CIBIO/InBIO-UP—Centro de Investigaç~ao em Biodiversidade e
Recursos Genéticos, University of Porto, Campus Agrario, Vair~ao, Portugal
Susana Martins University of Southern Denmark, Odense, Denmark
Barbara Mendes INEB—Instituto de Engenharia Biomédica, Universidade do
Porto, Porto, Portugal; Instituto de Investigaç~ao e Inovaç~ao em Saude, Universidade
do Porto, Porto, Portugal
José Augusto Guimar~ aes Morais iMed.UL, Faculty of Pharmacy, University of
Lisbon, Lisbon, Portugal
Rute Nunes INEB—Instituto de Engenharia Biomédica, University of Porto, Porto,
Portugal
Maria Beatriz P.P. Oliveira Requimte, Faculty of Pharmacy, University of Porto,
Porto, Portugal
Paulo Paix~
ao iMed.UL, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
Carla Pereira INEB—Instituto de Engenharia Biomédica, Universidade do Porto,
Porto, Portugal; FEUP—Faculdade de Engenharia da Universidade do Porto, Porto,
Portugal
Manuela Pintado CBQF—Centro de Biotecnologia e Química Fina—Laboratorio
Associado, Escola Superior de Biotecnologia, Universidade Catolica Portuguesa/
Porto, Porto, Portugal
Stephan Reichl Institut f€
ur Pharmazeutische Technologie, Technische Universit€at
Braunschweig, Braunschweig, Germany
Francisca Rodrigues Requimte, Faculty of Pharmacy, University of Porto, Porto,
Portugal; Fourmag Lda, Parque Industrial do Cruzeiro, Moreira de Conegos, Portugal
Ellaine Salvador Department of Anaesthesia and Critical Care, University of
Wurzburg, Wurzburg, Germany
Hélder A. Santos Division of Pharmaceutical Chemistry and Technology, Faculty
of Pharmacy, University of Helsinki, Helsinki, Finland
Bruno Sarmento Instituto de Investigaç~ao e Inovaç~ao em Saude, Universidade do
Porto, Porto, Portugal; INEB—Instituto de Engenharia Biomédica, Universidade do
Porto, Porto, Portugal; CESPU, Instituto de Investigaç~ao e Formaç~ao Avançada
em Ciências e Tecnologias da Saude, Instituto Superior de Ciências da Saude-Norte,
Departamento de Ciências Farmacêuticas, Gandra-PRD, Portugal
xiv List of contributors

Neha Shrestha Division of Pharmaceutical Chemistry and Technology, Faculty of


Pharmacy, University of Helsinki, Helsinki, Finland
Catia Silva CESPU, Instituto de Investigaç~ao e Formaç~ao Avançada em Ciências e
Tecnologias da Sa
ude, Gandra PRD, Portugal
Nuno Silva iMed.UL, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal

Natasa Skalko-Basnet University of Tromsø The Arctic University of Norway,
Tromsø, Norway
Alejandro Sosnik Technion-Israel Institute of Technology, Technion City, Haifa,
Israel
Fl
avia Sousa CESPU, Instituto de Investigaç~ao e Formaç~ao Avançada em Ciências
e Tecnologias da Sa
ude, Instituto Superior de Ciências da Saude-Norte, Gandra PRD,
Portugal
Ingunn Tho University of Oslo, Oslo, Norway
Ana Vanessa Nascimento University of Porto, Porto, Portugal; IINFACTS,
Instituto de Investigaç~ao e Formaç~ao Avançada em Ciências e Tecnologias da Saude,
CESPU, Cooperativa de Ensino Superior Politecnico e Universitario, Gandra PRD,
Portugal
Te
ofilo Vasconcelos BIAL, Portela & Ca , Trofa, Portugal; Instituto de Ciências
Biomédicas Abel Salazar, University of Porto, Porto, Portugal; Institute of Biomedical
Engineering (INEB), University of Porto, Porto, Portugal
List of figures

Figure 3.1.1 (a) Schematic representation of the three-dimensional oral mucosa.


(b) Closer view of different layers in the oral mucosa: epithelium,
fibroblasts, macrophages, capillaries, and extracellular matrix (ECM). 32
Figure 3.1.2 Filter-grown TR146 cells cultured submerged for 23 days.
Formalin-fixed paraffin-embedded section. Hematoxylin and eosin
staining demonstrates a stratified squamous epithelium-like tissue.
Original magnification 6400. Scale bar 10 mm. 34
Figure 3.1.3 Schematic representation of oral keratinocyte cultures on dead
de-epidermized dermis (DDED) supported by stainless steel grids and
filter. 36
Figure 3.2.1 Representation of the stomach mucosa showing its gastric glands
and gastric pit structures, as well as its different cell types. 42
Figure 3.3.1 Representation of the organization of the small intestine epithelium. 58
Figure 3.3.2 Sequential barriers during the transit of a drug molecule when orally
administered. 60
Figure 3.3.3 Schematic representation of the Transwell system. 62
Figure 3.3.4 Illustration of the Caco-2 monoculture model setup and culture
conditions. 63
Figure 3.3.5 Staining of co-culture day 3, Caco-2 and HT29-MTX single cultures
with alcian blue (which stains acidic mucosubstances) and eosin. 67
Figure 3.3.6 Illustration of the Caco-2/HT29-MTX co-culture model setup and
culture conditions. 68
Figure 3.3.7 Scanning electron microscopy analysis of a co-culture of Caco-2/Raji
B cells where M cells (M) were identified due to their lack of microvilli
in contrast to Caco-2 (C) cells. 68
Figure 3.3.8 Illustration of the Caco-2/Raji B co-culture model setup and
culture conditions. 69
Figure 3.3.9 Illustration of the Caco-2/HT29-MTX/Raji B co-culture model setup
and culture conditions. 70
Figure 3.3.10 Illustration of the 3D model setup comprising Caco-2, HT29-MTX,
fibroblasts, and THP-1 and culture conditions. 72
Figure 3.3.11 Assessment of the morphological and functional parameters to validate
in vitro intestinal models. 73
Figure 3.5.1 Schematic representation of absorption mechanisms in lung
epithelium. 102
Figure 3.5.2 Schematic representation of cellular culture (a), under submerged
conditions (b), and under an air–liquid interface (c). 106
xvi List of figures

Figure 3.6.1 The female genital tract: vagina, and ectocervix are composed of
stratified squamous epithelia and endocervix and uterus of single-layer
columnar epithelia. The transformation zone where two types of
epithelia meet is called the squamocolumnar junction. 116
Figure 3.6.2 Schematic description of the isolation and cultivation of human
ectocervical epithelia cells (hECE) from cervicovaginal tissue: (a) main
steps of the isolation procedure, (b) main steps of seeding on filter inserts. 118
Figure 3.6.3 Simple drawing of the permeability testing setup in the Transwell
system with epithelial cell membrane on the filter support. 120
Figure 3.6.4 Schematic representation of the dual-chamber model. 123
Figure 3.7.1 Schematic representation of the anatomy of the eye. 130
Figure 3.8.1 Morphology of human skin and different in vitro dermal permeation
models: (a) normal human skin epidermis, (b) EpiSkinÔ model,
(c) EpiDermÔ model, (d) StrataTestÔ, (e) SkinEthicÔ RHE model,
(f) EpiDerm FTÔ, and (g) EpiCSÔ (hematoxylin–eosin). 159
Figure 3.9.1 The cell associations at the BBB (Abbott, 2013). The NVU is a complex
cellular system that includes highly specialized ECs, a high concentration
of pericytes embedded in the EC basement membrane; astrocytic endfeet
associated with the parenchymal basement membrane, neurons, and
immune cells. 170
Figure 4.1.1 Scheme of Franz diffusion cell. After harvesting and preparing (c)
(tissue sample) and introducing (f) (magnetic stirrer), (a) (drug solution)
is introduced through the assembled diffusion chamber being retained
between (b) (donor compartment) and (d) (receiver compartment).
(d) should be filled with the receiver solution and preheated by inducing
the flow of water heated to 37  C through (e) (insulating jacket).
Periodically, samples may be taken from (g) (sampling port). 195
Figure 4.1.2 Scheme of Ussing diffusion cell. After harvesting and preparation, (c)
(tissue sample) is introduced in a proper compartment where the drug
solution will flow unidirectionally. (b) (Valve to adjust flow rate of
gassing) will allow control of the entrance and mixture of carbogenic
gas or nitrogen with the perfused solution in (a) (U-shaped glass tube).
(d) (Current injector) and (e) (voltmeter) allow TEER testing to assess
tissue viability. 196
Figure 4.2.1 Schematic representation of the small intestine and transport pathways
across the intestinal barrier: (a) transcellular passive diffusion,
(b) paracellular passive diffusion, (c–d) influx/efflux facilitated transport
by membrane proteins, (e) endocytosis with lysosome degradation,
and (f) transcytosis. 204
Figure 4.2.2 Schematic representation of the (a) classical Ussing chamber and
(b) Franz diffusion cell. 211
Figure 4.2.3 I. Microscopic image of Peyer’s patch (a) and non-Peyer’s patch (b) of
excised porcine tissue from the abattoir after transport to the laboratory.
Tissue deterioration was particularly evident in sections directly exposed to
the intestinal lumen. Tissue deterioration was expressed by destruction
of the functional epithelium and goblet cells layers (Ib) and by the presence
of cell agglomerates covering the remnants of villi (arrowheads). II.
Histological images of mucosal disintegration of porcine small intestine
(from freshly slaughtered piglets) at different time points and under dif-
ferent storage conditions. (a, b) Immediately after animal death; (c, d)
List of figures xvii

25 min after animal death; (c) stored at 0  C, (d) room temperature. Tissue
architecture was strongly affected before the permeability study began,
although normal viability values were obtained with the viability markers
used. (E, intestinal epithelium; LF, lymphoid follicle; PC, plica circularis;
TS, tela submucosa; VI, villi; arrowheads, mucosal disintegration.
Bar ¼ 100 mm). 218
Figure 4.2.4 Figure of an everted gut sac apparatus with dimensions (a) and
complete setup (b). 220
Figure 4.2.5 (a) Schematic representation of the medium-throughput system
(InTESTineÔ) developed by The Netherlands Organization for Applied
Scientific Research, in which ex vivo pig intestinal segments were
mounted. (b) Multiple devices can be arrayed at the same time. In the
figure, a 24-well setting is presented. 229
Figure 4.3.1 Butorphanol plasma levels after intravenous, intramuscular, and
nasal spray administration of a 2-mg dose. 239
Figure 4.3.2 Percent drug diffused through sheep nasal mucosa for curcumin
nanoemulsion and curcumin mucoadhesive nanoemulsion, using a
curcumin solution as reference. 247
Figure 4.3.3 In vitro ribavirin (RBV) permeation profiles across rabbit nasal mucosa
from an aqueous solution (empty circle, n ¼ 9) compared with ribavirin
powder (filled square, n ¼ 3). Data are expressed as mean  standard
error of the mean. 248
Figure 4.4.1 Morphology and thickness of epithelium in different lung regions. 257
Figure 4.4.2 Scheme of submucosal gland at the submucosa. 258
Figure 4.4.3 Schematic representation of IPL system. 262
Figure 4.5.1 The human vaginal mucosa of the fertile adult, presenting (E)
nonkeratinized stratified squamous epithelium and (LP) lamina propria
(H&E, 40). 275
Figure 4.5.2 The Gorodeski model. Effect of permeable supports on the stratification
of human ectocervical epithelial cells in vitro. Cells were cultured on
(a) solid or (b) filter supports. Cultures are shown at 12 days after
confluence. Arrowheads in (b) indicate attached envelopes (i.e., cells
after nuclei loss). Note that the envelopes have a more condensed
cytoplasm than the cells in the basal layer. The light areas between
the cells are intercellular spaces and extracellular matrix
(H&E, 400). 277
Figure 4.5.3 The EpiVaginalÔ model. H&E histological cross-sections of (a) VEC-100
tissue model containing normal human vaginal-ectocervical epithelial
cells, (b) human vaginal tissue explant, and (c) VEC-100-FT tissue
model. All tissues have nucleated basal and suprabasal cell layers
followed by layers in which nuclei are lost and cells become filled with
glycogen. 279
Figure 4.5.4 Reconstructed vaginal mucosa model developed by Sivard et al. as
observed by light microscopy after 14 days of submerged culture
on de-epidermized dermis. (a) 7 to 10 epithelial cell layers are seen
in a cryostat section stained with hematoxylin (scale bar ¼ 15 mm).
(b) Immunohistochemical labeling of Langerhans cells shows the
expression of langerin (in brown). Langerhans cells are present in the
basal and suprabasal layers of the mucosal epithelium (scale
bar ¼ 15 mm). 281
xviii List of figures

Figure 4.5.5 Vaginal mucosa reconstruction proposed by Bouschbacher et al. (2008)


and normal human vagina present similar morphology. (a) Schematic
representation of the various steps of vaginal mucosal reconstruction
(LC, Langerhans cells). (b and c) Immunohistochemical analysis of normal
human vagina and vaginal mucosa reconstructions. Collagen IV,
cytokeratin 13, involucrin, and loricrin expression is shown in (b) and
CD45 in (c). CD45þ cells are noted by black arrows. (d) Confocal
microscopy of Langerhans cells integrated in the vaginal mucosa recon-
struction. Langerhans cells precursors were labeled with CellTrackerÔ
Orange CMTMR (red) before seeding in the reconstructs. Nucleus were
labeled with 40 ,6-diamidino-2-phenylindole and reconstructs were subse-
quently analyzed by confocal microscopy. A top view of the mucosa
reconstruction is shown. Scale bars ¼ 20 mm. Open arrow shows the pol-
ycarbonate membrane containing 12 mm pores. 282
Figure 4.5.6 The HVEÔ model (H&E saffron staining). Image kindly provided by
SkinEthic/EpiSkin. 283
Figure 4.5.7 New Zealand rabbit vaginal histology. (a) Schematic diagram showing
a 10-week-old rabbit vagina after longitudinal vaginotomy. Photomicro-
graphs of cross-sections of the (b) upper, (c) middle, and (d) lower thirds
of the rabbit vagina (H&E, reduced from 40). Note that the muscle
component (arrowheads) is scarce but that sinusoidal structures (arrows)
are rich in the lower third. M, mucosal side; S, serosal side. 287
Figure 4.5.8 Histological variability of the vaginal mucosa of guinea pigs with various
phases of the estrous cycle (H&E). (a) Proestrus: the epithelium is
composed of a thick layer of mucous cells beneath which is a thinner layer
of immature stratified squamous epithelium; (b) estrus: the epithelium is
composed of a thick layer of mucous cells beneath which is a thick layer of
immature (keratinizing) stratified squamous epithelium; (c) metestrus:
mature stratified squamous epithelium overlies fibrous stroma of the
lamina propria; (d) early diestrus: immature stratified squamous epithelium
overlies fibrous stroma of the lamina propria; (e) late diestrus: many
mucous cells have developed in the stratified squamous epithelium;
(f) immature guinea pigs: stratified squamous epithelium. E, stratified
squamous epithelium; K, keratinizing epithelium; M, mucous cells;
S, fibrous stroma of the lamina propria. 288
Figure 4.5.9 Histological structure of sheep vaginal mucosa. H&E- and Masson’s
trichrome-stained sections showing the anterior and posterior vaginal
walls of (a–d) virgin, (e–h) parous, and (i–l) pregnant sheep. Asterisk
indicates blood vessels. Arrows in (g) indicate the three vaginal layers.
Scale bar ¼ 100 mm (H&E) or 250 mm (Masson’s trichrome). 290
Figure 4.5.10 Mean steady-state and mean estimated steady-state flux values of water,
17b-estradiol, r-arecoline, oxytocin, and vasopressin through human and
porcine vaginal mucosa. Hum, human vaginal mucosa; Por, porcine
vaginal mucosa. 292
Figure 4.5.11 Fluorescent confocal microscopy imaging of pig vaginal mucosa
obtained after incubation for 2 h with fluorescent nanoparticles. Green,
blue, and red signals are from nanoparticles, Hoechst 33342 (DNA), and
WGA, Alexa Fluor 594 conjugate (sialic acid/N-acetylglucosaminyl
residues at cell membranes/mucin), respectively. Scale bar ¼ 10 mm
and z-axis range is 6 mm. SE, sub-epithelium (lamina propria);
VL, vaginal lumen. 293
List of figures xix

Figure 4.5.12 Photomicrographs of porcine vaginal mucosa slices stained with H&E
technique and showing the morphological similarities (a and c) before
and (b and d) after freeze-thawing. Bar ¼ 100 mm for (a) and (b), 50 mm
for (c), and 30 mm for (d). 294
Figure 4.5.13 Photomicrographs of transverse sections of the anterior vaginal
mucosa of cows at different stages of the estrous cycle. (a) Proestrus
showing two to five layers of low columnar (L) to polyhedral (P) cells;
(b) estrus showing a single layer of tall, columnar mucus-producing
cells; (c) metestrus showing tall columnar mucus-secreting cells in one part
of the section (M) and several layers of polyhedral cells in another (P);
and (d) diestrus showing several layers of flattened epithelial cells
in some parts of the section (F) and several layers of polyhedral cells in
others (P). All sections stained with H&E, 260 in (a) and (b), 130 in (c)
and (d). 295
Figure 4.5.14 Comparative histology of the vaginal epithelium of rhesus and
pigtail macaques. (a) Technique used to measure epithelial thickness.
To eliminate observer bias, an electronic grid was placed on images
of vaginal epithelium and 16 measurements were taken at the
intersection of each vertical gridline and the basal lamina, taking two
measurements (thinnest and thickest) between each set of gridlines.
All measurements were taken at 100. Vaginal biopsies from the
same normal (b, c) pigtail or (d, e) rhesus macaque at day 0 (luteal
phase: b, d) and peak follicular (c, e) stage of the menstrual cycle.
Note that the keratinized layer (KE) is essentially absent in the luteal
phase of pigtail macaques. Also note that the epithelium is markedly
thinner in the luteal phase of both macaque species. 297
Figure 4.7.1 Representation of two possible routes for permeation through intact SC. 328
Figure 4.7.2 Schematic representation of Franz diffusion cell (a) and Keshary–Chien
diffusion cell (b). 334
Figure 4.7.3 Cumulative permeation patterns after finite and infinite dose
techniques. 338
Figure 4.8.1 Cell-based in vitro BBB models. (a) Monoculture. Endothelial
cells are grown on permeable insert. (b) Double co-culture.
Endothelial cells are grown on a permeable insert whereas astrocytes
are grown in the bottom of the well. (c) Triple co-culture. Endothelial
cells and astrocytes are grown on the upper and lower sides of the
permeable insert, respectively. Pericytes are grown in the bottom
of the well. 347
Figure 5.1 Relationship between the predicted logarithm of the effective human
jejunal permeability estimated by multiple linear regression and
in vivo observed values. The human jejunal permeability is predicted
by the equation log (Peff)(cm/h) ¼ 0.932 þ 0.763  log (Papp)(cm/h) þ
0.0324  RBN (Papp is determined in Caco-2 cells and RBN is
the number of rotatable bonds in the molecule) and has an
r ¼ 0.887. 359
Figure 5.2 Schematic representation of a whole-body PBPK model. Organs are
represented by compartments typically with well-stirred assumptions and
defined by physiologic volumes. Rate of drug arriving to the organ is
defined by both the drug concentration in blood and the specific blood
flow for each individual organ. Elimination organs may be defined by
either linear or saturable kinetics. 362
xx List of figures

Figure 5.3 Scheme of TAMD model. Dark blue compartments represent drug
particles in solid state; light blue compartments represent drug particles
dissolved. Stomach is represented by both states. Blue arrows represent
transit rates and its direction. Red arrows represent dissolution rate of
drug particles (Kd). Green arrows represent drug absorption rates and
vivid red arrows represent a sequenced drug transfer from enterocytes to
the portal vein, liver, and central compartment. Black arrows represent
drug transfer from central compartment into the exterior and into the
peripheral compartment, as well as from the peripheral into the central
compartment. 365
List of tables

Table 2.1 Different cell- and tissue-based in vitro models to predict drug permeability 5
Table 2.2 Summary of some physicochemical factors relevant for membrane
permeation by passive diffusion 15
Table 2.3 Equations used for permeability estimations in different methods 20
Table 3.1.1 Different types of oral mucosa with their properties 33
Table 3.2.1 Comparison of the anatomical lengths of the different organs along the
gastrointestinal tract and respective absorptive surface areas 47
Table 3.3.1 Alternatives to Caco-2 cells in drug permeability studies 65
Table 3.3.2 Cell morphology, features, and specific markers 74
Table 3.4.1 Atraumatic sampling methods to obtain human nasal epithelial cells 86
Table 3.4.2 Examples of techniques used to obtain viable primary nasal
epithelial cell cultures 89
Table 3.4.3 Example of protocol to culture serially passage nasal epithelial cells
using ALI and LCC conditions 94
Table 3.4.4 Examples of marker compounds for use in permeability studies 95
Table 3.4.5 Examples of drugs permeability studies using cell-based in vitro models 96
Table 3.5.1 Available models based on monocultures including cells of bronchial or
alveolar regions 105
Table 3.5.2 In vitro cell-based models using co-cultures of epithelial and other
cell lines (mainly cells from the immune, vascular systems, or support
cells) 105
Table 3.5.3 Molecular markers for alveolar epithelial type I and II cells 108
Table 3.6.1 Immortalized human cervicovaginal cell lines used in permeability studies 121
Table 3.7.1 List of the most relevant cell culture lines related to ocular tissues 136
Table 3.8.1 Three-dimensional models available on the market for dermal
permeation studies 160
Table 3.9.1 Qualitative comparison of principal BBB models 185
Table 4.1.1 Examples of permeability studies using porcine buccal mucosa as
tissue-based in vitro model 191
Table 4.2.1 Main factors governing intestinal permeability 207
Table 4.2.2 Comparison between in vitro methods versus in vivo assays,
and tissue-based versus cell-based 210
Table 4.2.3 Main advantages and drawbacks of Ussing chamber as a model for
drug permeability studies using excised tissues 212
Table 4.2.4 Parameters influencing experimental outcomes of diffusion chamber
assay that should be controlled and standardized 213
Table 4.2.5 Common buffers employed in drug permeability studies using intestinal
tissues and respective compositions 215
xxii List of tables

Table 4.2.6 Common markers used to assess viability and/or function of intestinal
tissues mounted in diffusion cells 216
Table 4.2.7 Electrical resistance of membrane and PD values reported in drug
permeability studies to fulfill criteria of viability of intestinal tissues
mounted in diffusion cells 217
Table 4.2.8 Factors affecting functionality and outcome of everted sac model 222
Table 4.3.1 Interspecies comparison of nasal cavity characteristics according to
conchae complexity 241
Table 4.3.2 Excised nasal tissue used to assess the intranasal route ex vivo 244
Table 4.4.1 Advantages and disadvantages of IPL 261
Table 4.5.1 Selected examples of permeability studies performed using
EpiVaginalÔ tissues 280
Table 4.5.2 Comparison between human and pig vaginal epithelium 291
Table 4.5.3 Selected examples of ex vivo permeability studies using human
cervicovaginal mucosa 298
Table 4.6.1 Summary of reported 3D cornea constructs for in vitro permeation
studies 312
Table 4.6.2 Established 3D cornea constructs not intended for permeation studies 315
Table 4.7.1 Guidelines for in vitro dermal absorption tests 331
Table 4.7.2 Parameters of in vitro dermal absorption tests, according
to detailed guidelines 332
Table 4.8.1 Immortalized cell lines 348
Introduction
Bruno Sarmento
1
Instituto de Investigaç~ao e Inovaç~ao em Saude, Universidade do Porto, Porto, Portugal;
INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal; CESPU,
Instituto de Investigaç~ao e Formaç~ao Avançada em Ciências e Tecnologias da Saude,
Instituto Superior de Ciências da Saude-Norte, Departamento de Ciências Farmacêuticas,
Gandra-PRD, Portugal

1.1 Introduction
Prediction of human drug absorption is a major goal in the design, optimization, and
selection of drugs and drug products intended for noninvasive delivery. There are
various techniques currently employed to evaluate the extent of drug absorption in
the different phases of drug discovery and development. Screening protocols to eval-
uate drug absorption include a range of preclinical methodologies, such as in silico, in
vitro, in situ, ex vivo, and in vivo.
Animal tests are the gold standard in preclinical studies to evaluate the absorption of
drugs, although they are time consuming and expensive and may not predict drug
behavior in humans. In vitro techniques for drug permeability assessment are less labo-
rious, cheaper, and more in line with the “three Rs” ethical policies (Polli, 2008).
Furthermore, in vitro models that thoroughly simulate conditions in the human mucosa
return absorption data that can help in selecting compounds entering into clinical
development. The impact of these models rests on their potential for rapid, cost-
effective, and adequate predictability of absorption potential in humans. In some cases,
the data are suitable to support regulatory filing of a new drug application.
Cell culture models offer the advantage of a highly defined tool in which the param-
eters and conditions can be easily changed. In addition, the use of human cell lines
avoids the kind of problems that arise when using animal tissue for in vitro experi-
ments (Sarmento et al., 2012).
Nevertheless, current in vitro methods often discard the proper integrity of the
simulated mucosae, are based on the confluence of cell monolayers, and do not take
into account the role of cellecell and celleextracellular stroma interactions on the ab-
sorption mechanisms. In vitro cell-based models have been optimized, becoming more
and more complex and incorporating more than one type of cell in order to more
closely resemble in vivo tissues. Therefore, new and improved tissue-based in vitro
models estimate drug absorption in a rapid, systematic, robust, and high-throughput
way to shorten drug development time.
Ex vivo tissue experiments, on the other hand, may represent an alternative
approach to track the fate of drugs across epithelial tissues, as far as tissue viability
and integrity is maintained. In fact, they would represent the closest to human condition.

Concepts and Models for Drug Permeability Studies. http://dx.doi.org/10.1016/B978-0-08-100094-6.00001-8


Copyright © 2016 Elsevier Ltd. All rights reserved.
2 Concepts and Models for Drug Permeability Studies

Considering all methodologies, the use of in vitro or ex vivo methods inherently


creates questions about the validity of extrapolating to the in vivo situation (Pelko-
nen, Boobis, & Gundert-Remy, 2001). In vitroein vivo correlation must allow the
prediction of the in vivo pharmacokinetics of a drug based on the in vitro drug
permeation profiles.
This book is intended to be an updated compilation of the most important buccal,
gastric, intestinal, pulmonary, nasal, vaginal, ocular, renal, skin, and bloodebrain bar-
rier in vitro models for predicting the permeability of drugs. The strategy of the book
will be based on three different approaches, summarizing the most recent achieve-
ments regarding models comprising immortalized cells with an intrinsic ability to
grow in a monolayer when seeded in permeable supports, primary cells isolated
from living organisms directly cultivating barrier monolayers, or tissue-based models
constructed with cell lines and extracellular matrix, resembling the tridimensional
structure of mucosae and other biological membranes.
Each model is described regarding the protocol of seeding and conservation, as well
as applications for specific drugs, taking into account the maintenance of physiologic
characteristics and functionality of epithelium, from the simplest immortalized cell-
based monoculture to the most complex engineered-tissue models.
Moreover, the equivalence between in vitro cell and tissue models and in vivo con-
ditions is discussed, highlighting how each model may provisionally resemble
different drug absorption routes.

References
Pelkonen, O., Boobis, A. R., & Gundert-Remy, U. (2001). In vitro prediction of gastrointestinal
absorption and bioavailability: an experts’ meeting report. European Journal of Clinical
Pharmacology, 57(9), 621e629.
Polli, J. (2008). In vitro studies are sometimes better than conventional human pharmacokinetic
in vivo studies in assessing bioequivalence of immediate-release solid oral dosage forms.
The AAPS Journal, 10(2), 289e299.
Sarmento, B., Andrade, F., da Silva, S. B., Rodrigues, F., das Neves, J., & Ferreira, D. (2012).
Cell-based in vitro models for predicting drug permeability. Expert Opinion on Drug
Metabolism & Toxicology, 8(5), 607e621.
Importance and applications
of cell- and tissue-based in vitro 2
models for drug permeability
screening in early stages of drug
development

Miguel Angel Cabrera-Pérez1,2, Marival Bermejo Sanz2, Victor Mangas Sanjuan2,
Marta Gonz 
alez-Alvarez 2
, Isabel Gonz 
alez Alvarez 2
1
Universidad Central Marta Abreu de Las Villas, Santa Clara, Villa Clara, Cuba;
2
Universidad Miguel Hernandez, San Juan de Alicante, Alicante, Espa~na

2.1 Introduction
Discovery, development, and registration of a new drug is a long, labor-intensive, risky,
and extremely costly process (DiMasi, Hansen, & Grabowski, 2003; Kola & Landis,
2004). One of the main reasons for the high attrition rates in drug development is the
number of new chemical entities with poor pharmacokinetic properties (Kubinyi, 2003).
The development of high-throughput biological screening (van de Waterbeemd,
2002), genomics, and combinatorial chemistry (Seneci & Miertus, 2000) has dramat-
ically increased the number of pharmacologically active molecules with unfavorable
biopharmaceutical properties. The main challenge for pharmaceutical researchers in
drug development is to identify the major pharmacokinetic hurdles of drug candidates
and validate the “developability” of a compound in the early stages to select superior
drug candidates with the best chances of market success (Stoner et al., 2004).
Currently, several in vitro experimental models with high-throughput capacity,
cost-effectiveness, and adequate predictability of absorption potential in humans are
available for evaluating intestinal permeability and transport across different biological
membranes (Sarmento et al., 2012).
Among the most popular in vitro models for assessing permeability/absorption are
those based in artificial membranes, such as parallel artificial membrane permeability
assay (PAMPA); systems based in cells, such as Caco-2, MDCK (Madin Darby canine
kidney), etc.; and systems based on tissues. Although all these methods do not reflect
the effect of physiological factors, their financial and ethical considerations have made
possible their successful use as decision-making tools during early drug development.
In this sense the goal of this chapter is to bring a general theoretical and practical
description of the different in vitro methods used to study drug permeability by
different administration routes.

Concepts and Models for Drug Permeability Studies. http://dx.doi.org/10.1016/B978-0-08-100094-6.00002-X


Copyright © 2016 Elsevier Ltd. All rights reserved.
4 Concepts and Models for Drug Permeability Studies

2.2 General considerations


Permeability data obtained from different experimental models can be different be-
tween laboratories, even though good in vitroein vivo correlations with human data
have been obtained. Among the main factors affecting the permeability values are
the laboratory materials, source of biological material, and nonstandardized intralabor-
atory and interlaboratory assays. In order to develop suitable permeability procedures,
several criteria are required, such as the experimental protocol should be confirmed by
standardized procedures, with good relationship with human data, and where high-
permeability internal standards, markers for tissue integrity and viability, and reference
compounds have to be used (Volpe, 2010).
During the drug development process many cell- and tissue-based in vitro models
have been used to study drug permeability through different administration routes.
Table 2.1 gives a general description of the most relevant experimental procedures
for permeability studies. A better description of each permeability assay will be pro-
vided over the course of this book.
Nevertheless, although in vivo animal permeability studies better reproduce the hu-
man results, the cell- and tissue-based in vitro models provide several advantages: (1) a
lesser amount of drug is needed for the assay; (2) few or no animals are used; (3) more
compounds can be screened; (4) mechanism of transport and metabolism can be stud-
ied; (5) the analytical evaluation is more simple compared with assays in biological
fluids; and (6) these in vitro models offers reproducibility, simplicity, and a reduced
operation cost. Finally, the application of in vitro models in drug permeability studies
represents a useful screening tool for assessing the biopharmaceutical appropriateness
of new chemical entities.

2.3 Drug transport


2.3.1 Transport mechanisms
A drug must interact with its receptors at the action sites or therapeutic targets for
exerting its pharmacological or toxic effects. The essential step, therefore, after drug
administration by any extravascular route is the absorption into the systemic circula-
tion to be distributed to tissues and organs.
The barriers that the drug has to cross before reaching the blood flow are integrated
by semipermeable cell membranes in all the administration routes. Drug transport
across a cellular barrier may involve transcellular (across the cell) or paracellular
(between cells) movement. While paracellular transport is size-restricted passive diffu-
sion, transcellular transport mechanisms include passive diffusion, carrier-mediated
transport (active or facilitated diffusion), and endocytosis.
Passive diffusion involves the movement of drug molecules down a concentra-
tion or electrochemical gradient without the expenditure of energy. This transport is
not saturable, does not involve a carrier, and shows a low structural specificity
Table 2.1 Different cell- and tissue-based in vitro models to predict drug permeability
Kind of
permeability Type of assay Tissue Cell Advantages Drawbacks

Importance and applications of cell- and tissue-based in vitro models


Buccal Diffusion cells • Isolated animal Similar to human tissue; low Limited surface area; tissue
(e.g., Franz) buccal tissue cost of acquisition; no damage by mastication;
compound loss due to laborious and time-
swallowing; resistance to consuming excision
irritation and loss of procedure; lack of blood
integrity (test many types flow; high variability;
of excipients); correlation large number of tissue
with in vivo animal data samples are needed; poor
prior to human evaluation potential for automation
• Hamster pouch Good correspondence to Culture growth rate,
epithelium human buccal mucosa number of differentiated
culture cell layers, lipid
• TR146 cell composition, and the
culture optimum days in culture
• EpiOral are not well
characterized; limited
number of compounds
tested
Intestinal Brush border From animal and human Nonspecific binding;
membrane tissue; useful for predictive only for
vesicles mechanistic studies passive transport
Artificial High-throughput screening Predictive only for passive
membranes capacity; low cost; transport; membrane
(PAMPA) different lipid retention for lipophilic
compositions; good compounds; dependency
predictability on the lipid membrane
composition and pH;
presence of a thicker

5
unstirred water layer
Continued
Table 2.1 Different cell- and tissue-based in vitro models to predict drug permeability—cont'd

6
Kind of
permeability Type of assay Tissue Cell Advantages Drawbacks

Isolated intestinal Animal or human origin; High intervariability


cells relatively easy to develop during cell preparation;
requires a radiolabeled
or fluorescent permeant

Cell culture models Caco-2 High screening capacity; Lack of mucus-secreting


derived from a human cells; thicker unstirred
colon adenocarcinoma; water layer and tighter
evaluation of transport monolayers compared to
mechanisms, absorption human small intestine;
enhancers, and toxicity; different expression of
reduction of laboratory metabolic enzymes; high
animals; the most inter- and intralaboratory

Concepts and Models for Drug Permeability Studies


extensively characterized permeability data; long
cell-based model; short differentiation period;
cell culture time; good low expression of uptake
potential for automation transporters; is not a
and miniaturization dynamic model: the
conditions of culture
influence the
performance of the cell
model
TC7 (Caco-2 Suitable for drug transport The expression of CYP3A
subclone) and biotransformation remains lower than the
studies; higher enzymatic level in human
expression enterocytes
MDCK (Madin The culture time to Nonhuman and
Darby canine confluence is shorter than nonintestinal origin;
kidney) Caco-2 cells; the
Another random document with
no related content on Scribd:
This eBook is for the use of anyone anywhere in the United
States and most other parts of the world at no cost and with
almost no restrictions whatsoever. You may copy it, give it away
or re-use it under the terms of the Project Gutenberg License
included with this eBook or online at www.gutenberg.org. If you
are not located in the United States, you will have to check the
laws of the country where you are located before using this
eBook.

1.E.2. If an individual Project Gutenberg™ electronic work is derived


from texts not protected by U.S. copyright law (does not contain a
notice indicating that it is posted with permission of the copyright
holder), the work can be copied and distributed to anyone in the
United States without paying any fees or charges. If you are
redistributing or providing access to a work with the phrase “Project
Gutenberg” associated with or appearing on the work, you must
comply either with the requirements of paragraphs 1.E.1 through
1.E.7 or obtain permission for the use of the work and the Project
Gutenberg™ trademark as set forth in paragraphs 1.E.8 or 1.E.9.

1.E.3. If an individual Project Gutenberg™ electronic work is posted


with the permission of the copyright holder, your use and distribution
must comply with both paragraphs 1.E.1 through 1.E.7 and any
additional terms imposed by the copyright holder. Additional terms
will be linked to the Project Gutenberg™ License for all works posted
with the permission of the copyright holder found at the beginning of
this work.

1.E.4. Do not unlink or detach or remove the full Project


Gutenberg™ License terms from this work, or any files containing a
part of this work or any other work associated with Project
Gutenberg™.

1.E.5. Do not copy, display, perform, distribute or redistribute this


electronic work, or any part of this electronic work, without
prominently displaying the sentence set forth in paragraph 1.E.1 with
active links or immediate access to the full terms of the Project
Gutenberg™ License.
1.E.6. You may convert to and distribute this work in any binary,
compressed, marked up, nonproprietary or proprietary form,
including any word processing or hypertext form. However, if you
provide access to or distribute copies of a Project Gutenberg™ work
in a format other than “Plain Vanilla ASCII” or other format used in
the official version posted on the official Project Gutenberg™ website
(www.gutenberg.org), you must, at no additional cost, fee or expense
to the user, provide a copy, a means of exporting a copy, or a means
of obtaining a copy upon request, of the work in its original “Plain
Vanilla ASCII” or other form. Any alternate format must include the
full Project Gutenberg™ License as specified in paragraph 1.E.1.

1.E.7. Do not charge a fee for access to, viewing, displaying,


performing, copying or distributing any Project Gutenberg™ works
unless you comply with paragraph 1.E.8 or 1.E.9.

1.E.8. You may charge a reasonable fee for copies of or providing


access to or distributing Project Gutenberg™ electronic works
provided that:

• You pay a royalty fee of 20% of the gross profits you derive from
the use of Project Gutenberg™ works calculated using the
method you already use to calculate your applicable taxes. The
fee is owed to the owner of the Project Gutenberg™ trademark,
but he has agreed to donate royalties under this paragraph to
the Project Gutenberg Literary Archive Foundation. Royalty
payments must be paid within 60 days following each date on
which you prepare (or are legally required to prepare) your
periodic tax returns. Royalty payments should be clearly marked
as such and sent to the Project Gutenberg Literary Archive
Foundation at the address specified in Section 4, “Information
about donations to the Project Gutenberg Literary Archive
Foundation.”

• You provide a full refund of any money paid by a user who


notifies you in writing (or by e-mail) within 30 days of receipt that
s/he does not agree to the terms of the full Project Gutenberg™
License. You must require such a user to return or destroy all
copies of the works possessed in a physical medium and
discontinue all use of and all access to other copies of Project
Gutenberg™ works.

• You provide, in accordance with paragraph 1.F.3, a full refund of


any money paid for a work or a replacement copy, if a defect in
the electronic work is discovered and reported to you within 90
days of receipt of the work.

• You comply with all other terms of this agreement for free
distribution of Project Gutenberg™ works.

1.E.9. If you wish to charge a fee or distribute a Project Gutenberg™


electronic work or group of works on different terms than are set
forth in this agreement, you must obtain permission in writing from
the Project Gutenberg Literary Archive Foundation, the manager of
the Project Gutenberg™ trademark. Contact the Foundation as set
forth in Section 3 below.

1.F.

1.F.1. Project Gutenberg volunteers and employees expend


considerable effort to identify, do copyright research on, transcribe
and proofread works not protected by U.S. copyright law in creating
the Project Gutenberg™ collection. Despite these efforts, Project
Gutenberg™ electronic works, and the medium on which they may
be stored, may contain “Defects,” such as, but not limited to,
incomplete, inaccurate or corrupt data, transcription errors, a
copyright or other intellectual property infringement, a defective or
damaged disk or other medium, a computer virus, or computer
codes that damage or cannot be read by your equipment.

1.F.2. LIMITED WARRANTY, DISCLAIMER OF DAMAGES - Except


for the “Right of Replacement or Refund” described in paragraph
1.F.3, the Project Gutenberg Literary Archive Foundation, the owner
of the Project Gutenberg™ trademark, and any other party
distributing a Project Gutenberg™ electronic work under this
agreement, disclaim all liability to you for damages, costs and
expenses, including legal fees. YOU AGREE THAT YOU HAVE NO
REMEDIES FOR NEGLIGENCE, STRICT LIABILITY, BREACH OF
WARRANTY OR BREACH OF CONTRACT EXCEPT THOSE
PROVIDED IN PARAGRAPH 1.F.3. YOU AGREE THAT THE
FOUNDATION, THE TRADEMARK OWNER, AND ANY
DISTRIBUTOR UNDER THIS AGREEMENT WILL NOT BE LIABLE
TO YOU FOR ACTUAL, DIRECT, INDIRECT, CONSEQUENTIAL,
PUNITIVE OR INCIDENTAL DAMAGES EVEN IF YOU GIVE
NOTICE OF THE POSSIBILITY OF SUCH DAMAGE.

1.F.3. LIMITED RIGHT OF REPLACEMENT OR REFUND - If you


discover a defect in this electronic work within 90 days of receiving it,
you can receive a refund of the money (if any) you paid for it by
sending a written explanation to the person you received the work
from. If you received the work on a physical medium, you must
return the medium with your written explanation. The person or entity
that provided you with the defective work may elect to provide a
replacement copy in lieu of a refund. If you received the work
electronically, the person or entity providing it to you may choose to
give you a second opportunity to receive the work electronically in
lieu of a refund. If the second copy is also defective, you may
demand a refund in writing without further opportunities to fix the
problem.

1.F.4. Except for the limited right of replacement or refund set forth in
paragraph 1.F.3, this work is provided to you ‘AS-IS’, WITH NO
OTHER WARRANTIES OF ANY KIND, EXPRESS OR IMPLIED,
INCLUDING BUT NOT LIMITED TO WARRANTIES OF
MERCHANTABILITY OR FITNESS FOR ANY PURPOSE.

1.F.5. Some states do not allow disclaimers of certain implied


warranties or the exclusion or limitation of certain types of damages.
If any disclaimer or limitation set forth in this agreement violates the
law of the state applicable to this agreement, the agreement shall be
interpreted to make the maximum disclaimer or limitation permitted
by the applicable state law. The invalidity or unenforceability of any
provision of this agreement shall not void the remaining provisions.
1.F.6. INDEMNITY - You agree to indemnify and hold the
Foundation, the trademark owner, any agent or employee of the
Foundation, anyone providing copies of Project Gutenberg™
electronic works in accordance with this agreement, and any
volunteers associated with the production, promotion and distribution
of Project Gutenberg™ electronic works, harmless from all liability,
costs and expenses, including legal fees, that arise directly or
indirectly from any of the following which you do or cause to occur:
(a) distribution of this or any Project Gutenberg™ work, (b)
alteration, modification, or additions or deletions to any Project
Gutenberg™ work, and (c) any Defect you cause.

Section 2. Information about the Mission of


Project Gutenberg™
Project Gutenberg™ is synonymous with the free distribution of
electronic works in formats readable by the widest variety of
computers including obsolete, old, middle-aged and new computers.
It exists because of the efforts of hundreds of volunteers and
donations from people in all walks of life.

Volunteers and financial support to provide volunteers with the


assistance they need are critical to reaching Project Gutenberg™’s
goals and ensuring that the Project Gutenberg™ collection will
remain freely available for generations to come. In 2001, the Project
Gutenberg Literary Archive Foundation was created to provide a
secure and permanent future for Project Gutenberg™ and future
generations. To learn more about the Project Gutenberg Literary
Archive Foundation and how your efforts and donations can help,
see Sections 3 and 4 and the Foundation information page at
www.gutenberg.org.

Section 3. Information about the Project


Gutenberg Literary Archive Foundation
The Project Gutenberg Literary Archive Foundation is a non-profit
501(c)(3) educational corporation organized under the laws of the
state of Mississippi and granted tax exempt status by the Internal
Revenue Service. The Foundation’s EIN or federal tax identification
number is 64-6221541. Contributions to the Project Gutenberg
Literary Archive Foundation are tax deductible to the full extent
permitted by U.S. federal laws and your state’s laws.

The Foundation’s business office is located at 809 North 1500 West,


Salt Lake City, UT 84116, (801) 596-1887. Email contact links and up
to date contact information can be found at the Foundation’s website
and official page at www.gutenberg.org/contact

Section 4. Information about Donations to


the Project Gutenberg Literary Archive
Foundation
Project Gutenberg™ depends upon and cannot survive without
widespread public support and donations to carry out its mission of
increasing the number of public domain and licensed works that can
be freely distributed in machine-readable form accessible by the
widest array of equipment including outdated equipment. Many small
donations ($1 to $5,000) are particularly important to maintaining tax
exempt status with the IRS.

The Foundation is committed to complying with the laws regulating


charities and charitable donations in all 50 states of the United
States. Compliance requirements are not uniform and it takes a
considerable effort, much paperwork and many fees to meet and
keep up with these requirements. We do not solicit donations in
locations where we have not received written confirmation of
compliance. To SEND DONATIONS or determine the status of
compliance for any particular state visit www.gutenberg.org/donate.

While we cannot and do not solicit contributions from states where


we have not met the solicitation requirements, we know of no
prohibition against accepting unsolicited donations from donors in
such states who approach us with offers to donate.

International donations are gratefully accepted, but we cannot make


any statements concerning tax treatment of donations received from
outside the United States. U.S. laws alone swamp our small staff.

Please check the Project Gutenberg web pages for current donation
methods and addresses. Donations are accepted in a number of
other ways including checks, online payments and credit card
donations. To donate, please visit: www.gutenberg.org/donate.

Section 5. General Information About Project


Gutenberg™ electronic works
Professor Michael S. Hart was the originator of the Project
Gutenberg™ concept of a library of electronic works that could be
freely shared with anyone. For forty years, he produced and
distributed Project Gutenberg™ eBooks with only a loose network of
volunteer support.

Project Gutenberg™ eBooks are often created from several printed


editions, all of which are confirmed as not protected by copyright in
the U.S. unless a copyright notice is included. Thus, we do not
necessarily keep eBooks in compliance with any particular paper
edition.

Most people start at our website which has the main PG search
facility: www.gutenberg.org.

This website includes information about Project Gutenberg™,


including how to make donations to the Project Gutenberg Literary
Archive Foundation, how to help produce our new eBooks, and how
to subscribe to our email newsletter to hear about new eBooks.

You might also like