throbber
SOLID-STATE PROPERTIES OF
`PHARMACEUTICAL MATERIALS
`
`STEPHEN R. BYRN
`GEORGE ZOGRAFI
`XIAOMING (SEAN) CHEN
`
`Gilead 2012
`I-MAK v. Gilead
`IPR2018-00126
`
`Page 1 of 55
`
`Copyright © 2017. John Wiley & Sons, Incorporated. All rights reserved.
`
`Byrn, Stephen, et al. Solid State Properties of Pharmaceutical Materials, John Wiley & Sons, Incorporated, 2017. ProQuest Ebook Central, http://ebookcentral.proquest.com/lib/nyli/detail.action?docID=4915572.
`Created from nyli on 2018-03-14 08:26:52.
`
`

`

`This edition first published 2017
`© 2017 John Wiley & Sons, Inc.
`
`All rights reserved. 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 or otherwise, except as permitted by law. Advice on how to obtain permission to reuse material from this title is
`available at http://www.wiley.com/go/permissions.
`
`The right of Stephen R. Byrn, George Zografi and Xiaoming (Sean) Chen to be identified as the author(s) of this work has been asserted in accordance
`with law.
`
`Registered Offices
`John Wiley & Sons, Inc., 111 River Street, Hoboken, NJ 07030, USA
`
`Editorial Office
`111 River Street, Hoboken, NJ 07030, USA
`
`For details of our global editorial offices, customer services, and more information about Wiley products visit us at www.wiley.com.
`
`Wiley also publishes its books in a variety of electronic formats and by print-on-demand. Some content that appears in standard print versions of this book
`may not be available in other formats.
`
`Limit of Liability/Disclaimer of Warranty
`The publisher and the authors make no representations or warranties with respect to the accuracy or completeness of the contents of this work and specifically
`disclaim all warranties; including without limitation any implied warranties of fitness for a particular purpose. This work is sold with the understanding that
`the publisher is not engaged in rendering professional services. The advice and strategies contained herein may not be suitable for every situation. In view of
`on-going research, equipment modifications, changes in governmental regulations, and the constant flow of information relating to the use of experimental
`reagents, equipment, and devices, the reader is urged to review and evaluate the information provided in the package insert or instructions for each chemical,
`piece of equipment, reagent, or device for, among other things, any changes in the instructions or indication of usage and for added warnings and precautions.
`The fact that an organization or website is referred to in this work as a citation and/or potential source of further information does not mean that the author or
`the publisher endorses the information the organization or website may provide or recommendations it may make. Further, readers should be aware that
`websites listed in this work may have changed or disappeared between when this works was written and when it is read. No warranty may be created or
`extended by any promotional statements for this work. Neither the publisher nor the author shall be liable for any damages arising here from.
`
`Library of Congress Cataloging-in-Publication Data:
`Names: Byrn, Stephen R., author. | Zografi, George, author. | Chen, Xiaoming (Sean), author.
`Title: Solid-state properties of pharmaceutical materials / Stephen R. Byrn, George Zografi,
`Xiaoming (Sean) Chen.
`Description: Hoboken, NJ : John Wiley & Sons, 2017. | Includes index.
`Identifiers: LCCN 2017005555 (print) | LCCN 2017008527 (ebook) | ISBN 9781118145302 (cloth) |
`ISBN 9781119264446 (Adobe PDF) | ISBN 9781119264453 (ePub)
`Subjects: LCSH: Solid state chemistry. | Solid dosage forms–Properties.
`Classification: LCC QD478 .B96 2017 (print) | LCC QD478 (ebook) | DDC 615.1028/4–dc23
`LC record available at https://lccn.loc.gov/2017005555
`
`Cover images: (Background) © Mimi Haddon/Getty images; (Inset images) Aeinleng, Nunchanit et al. “Physicochemical Performances of Indomethacin in
`Cholesteryl Cetyl Carbonate Liquid Crystal as a Transdermal Dosage.” AAPS PharmaSciTech 13.2 (2012): COVER. PMC
`Cover design by Wiley
`
`Set in 10/12pt TimesLTStd by Aptara Inc., New Delhi, India
`
`10 9 8 7 6 5 4 3 2 1
`
`Copyright © 2017. John Wiley & Sons, Incorporated. All rights reserved.
`
`Byrn, Stephen, et al. Solid State Properties of Pharmaceutical Materials, John Wiley & Sons, Incorporated, 2017. ProQuest Ebook Central, http://ebookcentral.proquest.com/lib/nyli/detail.action?docID=4915572.
`Created from nyli on 2018-03-14 08:26:52.
`
`Page 2 of 55
`
`

`

`1 S
`
`OLID-STATE PROPERTIES AND PHARMACEUTICAL
`DEVELOPMENT
`
`1.1
`
`INTRODUCTION
`
`Solid-state chemistry and the solid-state properties of phar-
`maceutical materials play an ever increasing and important
`role in pharmaceutical development. There is much more
`emphasis on physical characterization since the release of
`the International Committee on Harmonization (ICH) Q6A
`guidance on specifications. This guidance directs the scientist
`to determine what solid form is present in the drug substance
`(active pharmaceutical ingredient [API]) and drug product. It
`directs the manufacturer to “know what they have.” Addition-
`ally, the ICH Q8 guidance on development and the ICH Q9
`guidance on risk management require a firm understanding
`of how the medicine was developed and any risks involved.
`There are many more poorly soluble drugs under devel-
`opment. In many cases, the solid form of the API and
`the solid form and formulation in the drug product deter-
`mine apparent solubility that in turn determines blood lev-
`els. That is, the formulation determines bioavailability and
`therapeutic response. In these cases, it is even more impor-
`tant to physically characterize the API form and the formu-
`lations. Furthermore, the vast majority of medicines (drug
`products) are solids and those drug products that are not
`solids often start with solid APIs. In addition to solubil-
`ity and bioavailability, the solid form may affect stability,
`flow, compression, hygroscopicity, and a number of other
`properties.
`This book focuses on solid-state properties of pharmaceu-
`tical materials and methods of determining these properties.
`The authors have made every effort to include examples and
`
`case studies in order to illustrate the importance of knowing
`what you have. This book will focus on solid-state prop-
`erties and general strategies for physical characterization.
`Case studies and practical examples will be emphasized. In
`many respects, this book will illustrate that a medicine is
`more than a molecule. Additional goals include providing a
`full physical/analytical/operational definition of the different
`solid forms as well as other terms frequently used in phar-
`maceutical materials science including: polymorph, solvate,
`amorphous form, habit, nucleation, transformation, dissolu-
`tion, solubility, and stability.
`
`1.2 SOLID-STATE FORMS
`
`Pharmaceutical materials can exist in a crystalline or amor-
`phous state. Figure 1.1 illustrates the crystalline state as a
`perfectly ordered solid with molecules (circles) packed in an
`orderly array. Figure 1.1 illustrates an amorphous material
`as a disordered material with only short-range order. Crys-
`talline materials give an X-ray diffraction pattern because
`Bragg planes exist in the material (see Figure 1.2). Amor-
`phous materials do not give a diffraction pattern (Figure 1.2).
`Of course, there are many interesting cases where a phar-
`maceutical material shows an intermediate degree of order
`falling somewhere between the highly ordered crystalline
`state and the disordered amorphous state. From a thermody-
`namic point of view, crystalline materials are more stable but
`the rate of transformation of amorphous materials to crys-
`talline materials can be highly variable [1].
`
`Solid-State Properties of Pharmaceutical Materials, First Edition. Stephen R. Byrn, George Zografi and Xiaoming (Sean) Chen.
`© 2017 John Wiley & Sons, Inc. Published 2017 by John Wiley & Sons, Inc.
`
`Page 3 of 55
`
`1
`
`Copyright © 2017. John Wiley & Sons, Incorporated. All rights reserved.
`
`Byrn, Stephen, et al. Solid State Properties of Pharmaceutical Materials, John Wiley & Sons, Incorporated, 2017. ProQuest Ebook Central, http://ebookcentral.proquest.com/lib/nyli/detail.action?docID=4915572.
`Created from nyli on 2018-03-02 15:27:18.
`
`

`

`2
`
`SOLID-STATE PROPERTIES AND PHARMACEUTICAL DEVELOPMENT
`
`FIGURE 1.1
`Idealized view of crystalline (left panel) and amorphous (right panel) material. In
`this two-dimensional figure, the molecules are viewed as circles.
`
`Crystals of a pharmaceutical material from different
`sources can vary greatly in their size and shape. Typical parti-
`cles in different samples may resemble, for example, needles,
`rods, plates, and prisms. Such differences in shape are col-
`lectively referred to as differences in morphology. This term
`merely acknowledges the fact of different shapes. It does not
`distinguish among the many possible reasons for the different
`
`shapes. Naturally, when different compounds are involved,
`different crystal shapes would be expected as a matter of
`course. When batches of the same substance display crystals
`with different morphology, however, further work is needed
`to determine whether the different shapes are indicative of
`polymorphs, solvates, or just habits. Because these distinc-
`tions can have a profound impact on drug performance, their
`
`FIGURE 1.2 X-ray diffraction pattern of three samples, crystalline,
`amorphous.
`
`low crystallinity, and
`
`Page 4 of 55
`
`Copyright © 2017. John Wiley & Sons, Incorporated. All rights reserved.
`
`Byrn, Stephen, et al. Solid State Properties of Pharmaceutical Materials, John Wiley & Sons, Incorporated, 2017. ProQuest Ebook Central, http://ebookcentral.proquest.com/lib/nyli/detail.action?docID=4915572.
`Created from nyli on 2018-03-02 15:27:18.
`
`

`

`careful definition is very important to our discourse. At this
`time, only brief definitions are presented.
`
`r Polymorphs: When two crystals have the same chemical
`composition but different internal structure (molecular
`packing), they are polymorphic modifications, or poly-
`morphs (think of the three forms of carbon: diamond,
`graphite, and fullerenes). Polymorphs can result from
`different molecular packing, different molecular confor-
`mation, different tautomeric structure, or combinations
`of these.
`r Solvates: These crystal forms, in addition to containing
`molecules of the same given substance, also contain
`molecules of solvent regularly incorporated into a unique
`structure (think of wet, setting plaster: CaSO4 + 2H2O →
`⋅2H2O).
`CaSO4
`r Habits: Crystals are said to have different habits when
`samples have the same chemical composition and the
`same crystal structure (i.e., the same polymorph and unit
`cell) but display different shapes (think of snowflakes).
`
`Together, these solid-state physical modifications of a com-
`pound are referred to as crystalline forms. When differences
`between early batches of a substance are found by micro-
`scopic examination, for example, a reference to “form” is
`particularly useful in the absence of information that allows
`the more accurate description of a given variant batch (i.e.,
`polymorph, solvate, habit, or amorphous material). The term
`pseudopolymorphism is applied frequently to designate sol-
`vates. These solid-state modifications have different physical
`properties.
`
`SOLID-STATE FORMS
`
`3
`
`To put these important definitions into a practical con-
`text, we consider two cases (aspirin and flufenamic acid)
`in which a drug was crystallized from several different sol-
`vents and different-shaped crystals resulted in each exper-
`iment. Although sometimes dramatically different shapes
`were obtained upon changing solvents for the various crys-
`tallizations, the final interpretations in the two cases are dif-
`ferent. For aspirin, X-ray powder diffraction showed that all
`crystals regardless of shape had the same diffraction pattern.
`Thus, the different shaped crystals are termed crystal habits.
`For flufenamic acid, the different shaped crystals had differ-
`ent X-ray powder diffraction patterns. Subsequent analysis
`showed that the crystals did not contain solvent. Thus these
`different crystals are polymorphs.
`Further analysis of the crystals from this case provides
`the single crystal structure. The single crystal structure
`gives the locations of the atoms relative to a hypothetical unit
`cell. The unit cell is the smallest building block of a crys-
`tal. Figure 1.3 shows the unit cell of Form I of flufenamic
`acid. This unit cell contains four flufenamic acid molecules.
`Figure 1.4 shows a space-filling model of the contents of
`the flufenamic acid Form I unit cell. This figure illustrates
`Kitaigorodskii’s close-packing theory, which requires that
`the molecules pack to minimize free volume [2].
`Amorphous materials will be discussed in Chapter 6. In
`this introductory chapter as mentioned briefly above, amor-
`phous materials have no long range order and are thermody-
`namically metastable. An amorphous solid is characterized
`by a unique glass transition temperature Tg, the temperature
`at which it changes from a glass to a supercooled liquid or
`rubbery state. When T rises above Tg, the rigid solid can
`
`CF3
`
`HN
`
`COOH
`
`a
`
`0
`
`0
`
`c
`
`FIGURE 1.3 Single crystal structure the Form I polymorph of flufenamic acid (structure shown
`on the right panel).
`
`Page 5 of 55
`
`Copyright © 2017. John Wiley & Sons, Incorporated. All rights reserved.
`
`Byrn, Stephen, et al. Solid State Properties of Pharmaceutical Materials, John Wiley & Sons, Incorporated, 2017. ProQuest Ebook Central, http://ebookcentral.proquest.com/lib/nyli/detail.action?docID=4915572.
`Created from nyli on 2018-03-02 15:27:18.
`
`

`

`4
`
`SOLID-STATE PROPERTIES AND PHARMACEUTICAL DEVELOPMENT
`
`It is possible to make a “top 10” list of the differences
`between crystalline and amorphous materials. Crystalline
`materials have the following characteristics:
`
`1. higher purity,
`2. More physically and chemically stable, crystalline
`hydrate > anhydrous crystal > amorphous
`3. lower solubility,
`4. narrow and (usually) higher melting point range,
`5. harder,
`6. brittle – slip and cleavage,
`7. directionally dependent properties – anisotropy,
`8. less compressible,
`9. better flow and handling characteristics, and
`10. less hygroscopic.
`
`From this list, it is clear that crystalline materials are gen-
`erally more desirable unless they are so insoluble that they
`cannot be used as medicines.
`Not only do polymorphs show different X-ray powder
`diffraction patterns but they also have different unit cells,
`and different properties including thermal properties [6]. Fig-
`ure 1.5 shows the different crystal packing of the Forms I and
`II of sulfathiazole.
`Additionally, polymorphs are characterized as monotropic
`or enantiotropic depending upon their thermal properties
`[9, 10].
`
`r Monotropic polymorphs exist if the transition tem-
`perature between forms is greater
`than the melt.
`In monotropic polymorphs, one form is most stable
`throughout the temperature range.
`r Enantiotropic polymorphs exist if the transition tem-
`perature between forms occurs before melting. In this
`case, one form is more stable at one temperature. At a
`
`FIGURE 1.4 Space filling drawing of the unit cell of flufenamic
`acid Form I.
`
`flow and the corresponding increase in molecular mobility
`can result in crystallization or increased chemical reactiv-
`ity of the solid. Several historic papers describe some addi-
`tional details of amorphous materials. Pikal and coworkers at
`Eli Lilly showed that amorphous materials can have lower
`chemical stability [3], and Fukuoka et al. showed amorphous
`materials had a tendency to crystallize [4]. Nevertheless, in
`some cases, amorphous forms have been historically used
`as products. An excellent example is novobiocin [5], which
`exists in a crystalline and an amorphous form. The crys-
`talline form is poorly absorbed and does not provide thera-
`peutic blood levels; in contrast, the amorphous form is readily
`absorbed and is therapeutically active. Further studies show
`that the solubility rate of the amorphous form is 70 times
`greater than the crystalline form in 0.1 N HCl at 25◦C when
`particles <10 micron are used.
`
`FIGURE 1.5 Crystal packing and unit cells (grey) of Forms I (left panel) and II (right panel)
`of sulfathiazole. The grey and black molecules in Form I indicate two unrelated molecules in the
`asymmetric unit. Source: Kruger and Gafner, 1971 [7, 8]. Redrawn from data published.
`Page 6 of 55
`
`Copyright © 2017. John Wiley & Sons, Incorporated. All rights reserved.
`
`Byrn, Stephen, et al. Solid State Properties of Pharmaceutical Materials, John Wiley & Sons, Incorporated, 2017. ProQuest Ebook Central, http://ebookcentral.proquest.com/lib/nyli/detail.action?docID=4915572.
`Created from nyli on 2018-03-02 15:27:18.
`
`

`

`different temperature the other form is most stable. For
`flufenamic acid, Form I is most stable above the tran-
`sition temperature of 42◦C and Form III is most stable
`below the transition temperature. Practically, this means
`that slurrying at room temperature will convert Form I
`to Form III.
`
`Crystalline solvates contain solvents regularly incorporated
`into the crystal lattice. When the solvent is water the solid
`form is called a hydrate. Solvates and hydrates do not have
`the same composition as unsolvated materials. Solvates and
`hydrates are sometimes referred to as pseudopolymorphs or
`solvatomorphs. Interestingly, it is possible for solvates and
`hydrates to be polymorphic. In such a case one has polymor-
`phic solvates. Kuhnert Brandstatter in her 1971 book showed
`photomicrographs of 16 solvates of estradiol [11]. Figure 1.6
`shows the crystal structure of caffeine monohydrate. The
`crystal of caffeine is built up by stacking the layers shown in
`Figure 1.6 on top of each other. Thus the hydrate molecules
`are in tunnels in this solid form.
`It is important to note that the FDA (Food and Drug
`Administration) has defined polymorphs as “different crys-
`talline forms of the same drug substance. This may include
`solvation or hydration products (also known as pseudopoly-
`morphs) and amorphous forms. Per the current regulatory
`scheme, different polymorphic forms are considered the same
`active ingredients.” Thus, for purposes of registration, sci-
`entists are directed to define polymorphs more broadly to
`include amorphous forms, solvates, and hydrates.
`Cocrystals, that is, two component crystals, are another
`solid material of interest. Like solvates, the new crystalline
`
`SOLID-STATE FORMS
`
`5
`
`FIGURE 1.7 Crystal structure of a cocrystal (2-methoxy-4-
`nitrophenol-4-(dimethylamino) pyridine (2:1)). The unit cell param-
`eters are a = 6.880, b = 38.40, c = 8.454, and the space group is
`Pna21. Source: Burger and Ramberger, 1979 [9, 10]. Reproduced
`with the permission of Springer.
`
`structure imparts different properties including solubility,
`stability, and mechanical properties to the material. Of special
`interest are cocrystals with altered solubility or stability. Fig-
`ure 1.7 shows the crystal structure of a cocrystal of phenol and
`2-methoxy-4-nitrophenol–4-(dimethylamino)pyridine (2:1)
`[12]. The FDA has recently released a draft guidance defining
`cocrystals as “Solids that are crystalline materials composed
`of two or more molecules in the same crystal lattice.”
`Pharmaceutical salts are substances formed by a reaction
`of an acid and a base. The FDA has suggested the follow-
`ing definition of salts as “Any of numerous compounds that
`result from replacement of part or all of the acid hydrogen
`of an acid by a metal or a radical acting like a metal: an
`ionic or electrovalent crystalline compound. Per the current
`regulatory scheme, different salt forms of the same active
`moiety are considered different active ingredients.” When
`a carboxylic acid reacts with an amine a salt is typically
`formed (Scheme 1.1). However, the degree of proton transfer
`can vary depending on the acidity and basicity of the reacting
`groups. The FDA definition seems to encompass all of these
`materials.
`
`RCOOH + H2N − R′ ⟶ RCOO− ⋯ H3N+ − R
`
`FIGURE 1.6 Projection of the crystal structure of caffeine
`hydrate on the ab plane. Source: Burger and Ramberger, 1979
`[9, 10]. Reproduced with the permission of Springer.
`
`Page 7 of 55
`
`SCHEME 1.1
`
`Copyright © 2017. John Wiley & Sons, Incorporated. All rights reserved.
`
`Byrn, Stephen, et al. Solid State Properties of Pharmaceutical Materials, John Wiley & Sons, Incorporated, 2017. ProQuest Ebook Central, http://ebookcentral.proquest.com/lib/nyli/detail.action?docID=4915572.
`Created from nyli on 2018-03-02 15:27:18.
`
`

`

`6
`
`SOLID-STATE PROPERTIES AND PHARMACEUTICAL DEVELOPMENT
`
`FIGURE 1.8 Crystal packing of calcium tolfenamate trihydrate showing hydrogen bonding net-
`work. The directions of the unit cell axis are (a) vertical, (c) across, and (b) out of the plane of the
`paper. Source: Atassi, 2007 [13]. Reproduced with the permission of Purdue University.
`
`Figure 1.8 shows the crystal structure of calcium tolfe-
`namate trihydrate. It is clear that the unit cell is composed
`of regions containing mostly hydrocarbon functional groups
`and regions containing polar functionalities. This type of
`crystal packing is typical for salts.
`
`1.3
`
`ICH Q6A DECISION TREES
`
`In 1995, Byrn et al. from Purdue University and the FDA pub-
`lished a paper using decision trees to describe a strategy to
`identify the best solid form early in development. In this way,
`it is possible to ensure uniformity of solid form in clinical
`trials and resolve solid-state issues before critical stages of
`development. The decision trees also suggested appropriate
`analytical methods for control. Four decision trees were pre-
`sented: polymorphs, hydrates/solvates, desolvated solvates,
`and amorphous forms [14].
`In the late 1990s, the ICH used a similar decision tree
`approach to describe how specification for the solid form
`in drug substances (API) and drug product should be deter-
`mined. Several decision trees were presented in the ICH
`Q6A document including decision trees on particle size and
`polymorphs. The ICH utilized the broadened definition of
`polymorphs that includes hydrates, solvates, and amorphous
`
`forms. The ICH decision trees are divided into three ques-
`tions as shown in Figures 1.9–1.11.
`These three decision trees outline a strategy that is widely
`used during drug development. Most firms conduct an early
`polymorph screen to address question number 1. Once new
`forms have been identified, they are physically characterized
`(solubility, stability, melting point) and an effort is made to
`understand whether these differences in properties will affect
`drug product safety, performance, or efficacy. If the different
`solid forms can affect safety, efficacy, or performance then
`question 3 is addressed by determining whether drug product
`testing can detect changes in ratios of these forms. Addition-
`ally, the ratios of forms are monitored during stability studies
`to make sure changes that affect performance, safety, or effi-
`cacy do not occur. Using this strategy, it is possible to find
`the best solid form for development rapidly.
`
`“BIG QUESTIONS” FOR DRUG
`1.4
`DEVELOPMENT
`
`In addition to selecting the solid form, Table 1.1 lists other
`critical issues/measurements required for drug development.
`Another way to think about drug development is to think
`of this process in terms of answering a series of questions
`we call the “Big Questions.” These must be answered to be
`
`Page 8 of 55
`
`Copyright © 2017. John Wiley & Sons, Incorporated. All rights reserved.
`
`Byrn, Stephen, et al. Solid State Properties of Pharmaceutical Materials, John Wiley & Sons, Incorporated, 2017. ProQuest Ebook Central, http://ebookcentral.proquest.com/lib/nyli/detail.action?docID=4915572.
`Created from nyli on 2018-03-02 15:27:18.
`
`

`

`“BIG QUESTIONS” FOR DRUG DEVELOPMENT
`
`7
`
`FIGURE 1.9
`ICH Q6A question 1 on polymorphs: Can different polymorphs be formed? Source:
`ICH Harmonized Tripartite Guideline, 1999. Reproduced with the permission of ICH.
`
`FIGURE 1.10
`ICH Q6A question 2 on polymorphs: Do the forms have different properties (sol-
`ubility, stability, melting point)? Source: ICH Harmonized Tripartite Guideline, 1999. Reproduced
`with the permission of ICH.
`
`Copyright © 2017. John Wiley & Sons, Incorporated. All rights reserved.
`
`Byrn, Stephen, et al. Solid State Properties of Pharmaceutical Materials, John Wiley & Sons, Incorporated, 2017. ProQuest Ebook Central, http://ebookcentral.proquest.com/lib/nyli/detail.action?docID=4915572.
`Created from nyli on 2018-03-02 15:27:18.
`
`Page 9 of 55
`
`

`

`8
`
`SOLID-STATE PROPERTIES AND PHARMACEUTICAL DEVELOPMENT
`
`FIGURE 1.11
`ICH Q6A question 3 on polymorphs: Does products performance testing provide
`adequate control I polymorph ratio changes (e.g., dissolution)? Source: ICH Harmonized Tripartite
`Guideline, 1999. Reproduced with the permission of ICH.
`
`able to develop a drug to clinical trials and beyond. These
`questions are as follows:
`
`r What is the structure of the compound?
`r What is the likely dose?
`r What is the route of administration and desired dosage
`form?
`r What is the indication?
`r How difficult is it to synthesize?
`r How soluble is the compound/formulation?
`r How well is the compound absorbed? What is its BCS
`(Biopharmaceutical Classification System) class?
`r What is the toxicology of the compound? What is its
`NOAEL (no-observed-adverse-effect-level)? What is its
`MTD (maximum tolerated dose)?
`
`Page 10 of 55
`
`TABLE 1.1 Critical Issues for Drug Development
`
`Polymorph selection
`Chemical synthesis
`Salt selection (optional)
`Assay/Impurities
`Particle size
`Physical characterization and properties
`Dissolution/solubility
`Consistency
`Stability
`Validated methods/processes
`Regulatory issues
`Intellectual property
`
`Copyright © 2017. John Wiley & Sons, Incorporated. All rights reserved.
`
`Byrn, Stephen, et al. Solid State Properties of Pharmaceutical Materials, John Wiley & Sons, Incorporated, 2017. ProQuest Ebook Central, http://ebookcentral.proquest.com/lib/nyli/detail.action?docID=4915572.
`Created from nyli on 2018-03-02 15:27:18.
`
`

`

`r What biomarkers are available to monitor clinical trials?
`r What doses should be used for Phase 2 clinical trial?
`r What are its solid-state properties and physical charac-
`terization?
`r How chemically stable is the compound?
`r How physically stable is the compound?
`r How well will the powder flow?
`r Is moisture an issue?
`r What is the design, composition, and manufacturing
`procedure of the formulation/product?
`
`As has already been discussed, the solubility of the compound
`is a critical quality attribute important for specifications and
`development. The solubility of a solid substance is the con-
`centration at which the solution phase is in equilibrium with
`a given solid phase at a stated temperature and pressure.
`Under these conditions, the solid is neither dissolving nor
`continuing to crystallize. Note that the definition implies the
`presence of a specific solid phase. Once determined under the
`stated conditions, however, we can talk about the “solubility”
`of a given phase (e.g., a specific polymorph or pseudopoly-
`morph) as a quantity, even in the absence of that solid phase.
`Use of the term “equilibrium” in connection with crystalliz-
`ing systems requires clarification. When a substance exists in
`more than one crystal form, that is, when other polymorphs
`are possible, only the least soluble of these at a given tem-
`perature is considered the most physically stable form at that
`temperature, all others are considered to be metastable forms.
`In given cases, a solution of a substance may be in apparent
`equilibrium with one of these metastable phases for a long
`time, in which case, the system is in metastable equilibrium
`and is expressing the thermodynamic solubility of that solid
`form.
`It is important to stress the difference between poly-
`morphs and solvates (pseudopolymorphs) at this point. If
`a solvate/pseudopolymorph exists, it is always (with few
`exceptions) the most stable form in the solvent that produces
`the pseudopolymorph.
`Undersaturation pertains to solutions at a lower concentra-
`tion than the saturation value (i.e., diluted solutions). Crystals
`will dissolve in undersaturated solutions. Saturation is the
`state of a system where the solid is in equilibrium with the
`solution, or in other words, the solution will neither dissolve
`crystals nor let them grow (i.e., the concentration of the solu-
`tion represents the solubility value for that crystalline phase).
`Supersaturation pertains to solutions that, for one reason or
`another (e.g., rapid cooling of a saturated solution without
`forming crystals), are at a higher concentration than the satu-
`ration value. Supersaturation is required for crystals to grow.
`The solubility and permeability are combined to deter-
`mine the BCS class (Table 1.2). BCS class I drugs dis-
`solve easily and are easily transported into the blood stream
`
`ACCELERATING DRUG DEVELOPMENT
`
`9
`
`TABLE 1.2 Biopharmaceutical Classification System (BCS)
`
`BCS Classification
`
`Solubility
`
`Permeability
`
`BCS class I
`BCS class II
`BCS class III
`BCS class IV
`
`High
`Low
`High
`Low
`
`High
`High
`Low
`Low
`
`because they are highly permeable with respect to the mem-
`branes in the gastrointestinal (GI) tract. BCS class III and
`IV drug have poor permeability and are generally difficult
`to develop. BCS class II drugs are of the greatest interest to
`pharmaceutical scientists because the structure of the solid,
`the formulation, its physical character, and many other fac-
`tors are likely to have a significant effect on bioavailabil-
`ity and ultimately safety, performance, and efficacy. Several
`important drugs that are widely prescribed are BCS Class
`II including atorvastatin calcium, celecoxib, efavirenz, irbe-
`sartan, lopinavir, medroxyprogesterone acetate, raloxifene
`hydrochloride, simvastatin, and warfarin sodium. Of the mar-
`keted drugs nearly 70% are in BCS Class I or II with 31%
`being in BCS Class II. It has been estimated that as high as
`80% of the drugs under development are BCS Class II.
`
`1.5 ACCELERATING DRUG DEVELOPMENT
`
`Accelerating drug development has been a goal of pharma-
`ceutical scientists for many years. In 1995, Colin Gardner of
`Merck introduced a flow chart showing synthesis of the API
`and development of clinical supplies for first in human trials
`in 1 year. In this early flow chart, drug substance synthesis
`and process development were carried out in parallel with
`preformulation/formulation design/development and safety
`studies. Despite the early introduction of the concept that an
`IND (investigational new drugs) can be submitted in 1 year,
`it has been difficult to achieve this goal except in very favor-
`able cases. One of the difficulties is the availability of API,
`and another one of the difficulties is accelerating toxicology
`studies.
`IND-I-GO
`introduced their
`In 2007, Aptuit/SSCI
`(INDIGO) program offering fast development in a Contract
`Research Organization (CRO) environment. INDIGO was
`tailored to working with BCS Class II compounds and poorly
`soluble compounds. This INDIGO offering was supported
`by an example case study on the poorly soluble drug itra-
`conazole and was summarized in a recent publication [15].
`Additionally, Byrn et al. and Byrn and Henck outlined strate-
`gies based on solid-state chemistry for reducing development
`time [16,17]. These publications contained much more detail
`on how to carry out screens and are discussed in more detail
`below. In this same timeframe, Chorus a Lilly-based firm
`focused on fast development introduced their strategy for
`
`Page 11 of 55
`
`Copyright © 2017. John Wiley & Sons, Incorporated. All rights reserved.
`
`Byrn, Stephen, et al. Solid State Properties of Pharmaceutical Materials, John Wiley & Sons, Incorporated, 2017. ProQuest Ebook Central, http://ebookcentral.proquest.com/lib/nyli/detail.action?docID=4915572.
`Created from nyli on 2018-03-02 15:27:18.
`
`

`

`10
`
`SOLID-STATE PROPERTIES AND PHARMACEUTICAL DEVELOPMENT
`
`accelerated development to Phase II [18]. In this early pub-
`lication, they suggested that it was possible to develop a
`compound to Phase II in 30 months for $3 million dollars in
`contrast to the industry average of 42 months and $30 million
`dollars. The Chorus approach involves a virtual company that
`heavily uses preferred CROs. Chorus has been quite success-
`ful, and Lilly has now established Chorus as an independent
`entity.
`In this same timeframe, PricewaterhouseCoopers intro-
`duced a concept of limited launch with what they have termed
`a “live license” that permits a company to market a drug under
`very restricted conditions. In one manifestation of this con-
`cept, they suggested launch of a drug after 1.5 years. The
`details of this strategy are not clear, but it appears that the
`proposal involves intr

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket