throbber
A Practical GuidefreomCandidate Drug
`
`Pharmaceutical
`Preformulation and
`Formulation
`
`SelectoFgetealommere al Dosage Form
`
`IPR2018-00126
`
`Page 1 of 610
`
`I-MAK 1020
`
`

`

`1408_FM 6/10/03 10:46 AM Page i
`
`PHARMACEUTICAL
`PREFORMULATION
`AND
`FORMULATION
`
`A Practical Guide from
`Candidate Drug Selection to
`Commercial Dosage Form
`
`Mark Gibson
`Editor
`
`IHSfi Health Group
`
`An IHSfi GROUP Company
`
`Your Enterprise Solution to (cid:31)
`Global Healthcare Knowledge
`
`IPR2018-00126
`
`Page 2 of 610
`
`I-MAK 1020
`
`

`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`disclaimer Page 1 Monday, June 9, 2003 12:22 PM
`
`Library of Congress Cataloging-in-Publication Data
`
`Catalog record is available from the Library of Congress
`
`This book contains information obtained from authentic and highly regarded sources. Reprinted material is quoted with
`permission, and sources are indicated. A wide variety of references are listed. Reasonable efforts have been made to publish
`reliable data and information, but the authors and the publisher cannot assume responsibility for the validity of all materials
`or for the consequences of their use.
`
`Neither this book nor any part may be reproduced or transmitted in any form or by any means, electronic or mechanical,
`including photocopying, microfilming, and recording, or by any information storage or retrieval system, without prior
`permission in writing from the publisher.
`
`The consent of CRC Press LLC does not extend to copying for general distribution, for promotion, for creating new works,
`or for resale. Specific permission must be obtained in writing from CRC Press LLC for such copying.
`
`Direct all inquiries to CRC Press LLC, 2000 N.W. Corporate Blvd., Boca Raton, Florida 33431.
`
`Trademark Notice:
`Product or corporate names may be trademarks or registered trademarks, and are used only for
`identification and explanation, without intent to infringe.
`
`Visit the CRC Press Web site at www.crcpress.com
`
`© 2004 by Interpharm/ CRC
`
`No claim to original U.S. Government works
`Library of Congress Card Number 1-57491-120-1
`Printed in the United States of America 1 2 3 4 5 6 7 8 9 0
`Printed on acid-free paper
`
`IPR2018-00126
`
`Page 3 of 610
`
`I-MAK 1020
`
`

`

`1408_FM 6/10/03 10:46 AM Page iii
`
`CONTENTS
`
`Preface
`
`Contributors
`
`1.
`
`Introduction and Perspective
`
`Mark Gibson
`
`Drug Development Drivers, Challenges, Risks and Rewards
`Current Trends in the Pharmaceutical Industry
`Lessons Learnt and the Way Forward
`Scope of the Book
`References
`
`2.
`
`3.
`
`PART I: Aiding Candidate Drug Selection
`Aiding Candidate Drug Selection:
`Introduction and Objectives
`
`Mark Gibson
`
`Stages of the Drug Discovery and Development Process
`Summary
`References
`
`Preformulation Predictions from Small Amounts of Compound
`as an Aid to Candidate Drug Selection
`Gerry Steele
`
`Initial Physicochemical Characterization
`Initial Solubility
`Initial Stability Investigations
`Crystallinity
`Crystal Morphology
`Hygroscopicity
`Salt Selection
`Methods for Evaluating Physicochemical Properties
`Concluding Remarks
`Acknowledgements
`References
`
`vii
`
`ix
`
`1
`
`2
`6
`8
`10
`11
`
`15
`
`15
`20
`20
`
`21
`
`22
`28
`34
`41
`46
`48
`49
`58
`87
`88
`88
`
`iii
`
`IPR2018-00126
`
`Page 4 of 610
`
`I-MAK 1020
`
`

`

`1408_FM 6/10/03 10:46 AM Page iv
`
`iv
`
`4.
`
`Pharmaceutical Preformulation and Formulation
`
`Biopharmaceutical Support in Candidate Drug Selection
`Anna-Lena Ungell and Bertil Abrahamsson
`
`Drug Dissolution and Solubility
`Luminal Interactions
`Absorption/Uptake over the GI Membranes
`Models for Studying the Absorption Potential of Drugs
`Permeability Coefficients versus Fa
`In Vivo Techniques for Studies in Man
`Vehicles for Absorption Studies
`Functional Use of Absorption Models
`References
`
`PART II: Early Drug Development
`Early Drug Development: Product Design
`Mark Gibson
`
`5.
`
`The Importance of Product Design
`Product Design Considerations
`Concluding Remarks
`References
`
`6.
`
`Preformulation as an Aid to Product Design in Early Drug Development
`Gerry Steele
`
`Solid Dosage Forms
`Solution Formulations
`Freeze-Dried Formulations
`Suspensions
`Topical/Transdermal Formulations
`Inhalation Dosage Forms
`Compatibility
`References
`
`7.
`
`Biopharmaceutical Support in Formulation Development
`Bertil Abrahamsson and Anna-Lena Ungell
`
`In Vitro Dissolution
`Bioavailability Studies
`In Vitro/In Vivo Correlations
`Animal Models
`Imaging Studies
`References
`
`97
`
`100
`111
`117
`118
`134
`135
`139
`141
`143
`
`157
`
`157
`158
`173
`173
`
`175
`
`175
`196
`210
`214
`215
`217
`223
`228
`
`239
`
`241
`257
`269
`276
`279
`289
`
`PART III: From Product Design to Commercial Dosage Form
`Product Optimisation
`295
`
`8.
`
`Product Optimisation Purpose and Scope
`Excipient and Pack Optimisation Considerations
`
`Mark Gibson
`
`295
`296
`
`IPR2018-00126
`
`Page 5 of 610
`
`I-MAK 1020
`
`

`

`1408_FM 6/10/03 10:46 AM Page v
`
`Sources of Information
`Expert Systems
`Experimental Design
`Stability Testing
`Developing Specifications
`Process Design, Process Optimisation and Scale-Up
`Validation and Launch
`Acknowledgements
`References
`
`9.
`
`Parenteral Dosage Forms
`
`Joanne Broadhead
`
`Guiding Principles for Simple Parenteral Solutions
`Choice of Excipients
`Sterility Considerations
`Strategies for Formulating Poorly Soluble Drugs
`Strategies for Formulating Unstable Molecules
`Strategies for the Formulation of Macromolecules
`Liposomal Delivery Systems
`Sustained-Release Parenteral Formulations
`In Vitro and In Vivo Testing Methods
`Packaging of Parenteral Products
`Manufacturing of Parenteral Products
`Administration of Parenteral Products
`Parenteral Products and the Regulatory Environment
`References
`
`10.
`
`Inhalation Dosage Forms
`
`Paul Wright
`
`Lung Deposition
`Particle Sizing
`Dry Powder Inhalers
`Metered Dose Inhalers
`Nebulisers
`Standards
`Future
`References
`Bibliography
`
`11. Oral Solid Dosage Forms
`
`Peter Davies
`
`Powder Technology
`Powder Flow
`Mixing
`Compaction
`Solid Dosage Forms
`Tablets
`Hard Gelatin Capsules
`
`Contents
`
`v
`
`304
`305
`309
`313
`316
`319
`323
`327
`327
`
`331
`
`332
`334
`336
`336
`340
`342
`343
`343
`346
`347
`348
`350
`351
`353
`
`355
`
`356
`357
`361
`364
`372
`374
`375
`376
`378
`
`379
`
`381
`382
`388
`390
`403
`403
`441
`
`IPR2018-00126
`
`Page 6 of 610
`
`I-MAK 1020
`
`

`

`1408_FM 6/10/03 10:46 AM Page vi
`
`vi
`
`Pharmaceutical Preformulation and Formulation
`
`Soft Gelatin Capsules
`Summary
`References
`
`12. Ophthalmic Dosage Forms
`
`Mark Gibson
`
`Ocular Topical Drug Delivery Issues and Challenges
`Drug Candidate Selection
`Product Design Considerations
`Product Optimisation Considerations
`Processing Considerations
`Concluding Remarks
`References
`
`13. Aqueous Nasal Dosage Forms
`
`Nigel Day
`
`Nasal Anatomy and Physiology
`Formulation Selection Considerations
`Device Selection Considerations
`Regulatory Aspects
`Special Considerations for Peptide Nasal Delivery
`References
`Additional Reading
`
`14.
`
`Topical and Transdermal Delivery
`Kenneth A. Walters and Keith R. Brain
`
`The Skin and Percutaneous Absorption
`Drug Candidate Selection and Preformulation
`Formulation
`Concluding Remarks
`Bibliography
`References
`
`Index
`
`453
`455
`456
`
`459
`
`460
`464
`465
`473
`482
`486
`488
`
`491
`
`494
`496
`499
`506
`508
`511
`513
`
`515
`
`516
`534
`543
`567
`567
`569
`
`581
`
`IPR2018-00126
`
`Page 7 of 610
`
`I-MAK 1020
`
`

`

`1408_FM 6/10/03 10:46 AM Page vii
`
`PREFACE
`
`In my industrial career, I have gained broad experience working on many different pharma-
`ceutical development projects and dosage forms, spanning candidate drug selection through
`technology transfer to production and subsequent launch. With this background and with
`Gerry Steele’s encouragement and rich experience in preformulation studies, I planned a
`book that would emphasize what practical studies need to be undertaken, for what reasons
`and during what key stages of the drug development process. In addition to preformulation,
`I considered it essential to include biopharmaceutics (an area of emerging importance) as
`well as formulation aspects. I soon realized I could not commit the time to writing all the
`chapters in such a book, and, indeed, I considered the book would be more comprehensive by
`involving other contributors and adding their special knowledge and experiences.
`I am extremely grateful to all the contributors in this book, who have given up so much
`of their time to create their specialised chapters. I would specially like to thank Anna-Lena
`Ungell and Bertil Abrahammson for the two biopharmaceutics chapters, and also Jo Broad-
`head for stepping in at the final hour and writing the chapter on parenterals. Without these
`chapters, the book would have been incomplete.
`In the past few years, I have written internal company guidelines and given seminars ex-
`ternally on a logical approach to product development and technology transfer. This ap-
`proach emphasises the importance of starting development of an NCE with product design
`prior to commencing product and process optimisation, scale-up and technology transfer.
`Two people I would particularly like to thank for their input and support for these concepts
`are Gordon France and Mike Dey. This logical approach to ensure that products are devel-
`oped efficiently and moved speedily to market is part of my own contribution to the chapters
`in this book.
`Preparing this book took longer than anticipated, and it contains more pages than ex-
`pected. I have many vivid memories of one-finger typing on my laptop in airport lounges and
`hotel rooms and at home late in the evenings or on weekends. While my typing has become
`considerably faster, I hasten to add that I have progressed only to two-finger typing! I am in-
`debted to my secretarial support, particularly Hayley Pruden, who has much better typing
`skills than I, to complete the figures and tables for this book.
`
`vii
`
`IPR2018-00126
`
`Page 8 of 610
`
`I-MAK 1020
`
`

`

`1408_FM 6/10/03 10:46 AM Page viii
`
`viii
`
`Pharmaceutical Preformulation and Formulation
`
`This book should prove to be a useful guide to practitioners working in the pharmaceu-
`tical industry, including R&D scientists, technicians and managers. In the words of Francis
`Bacon (1561–1626):
`
`Some books are to be tasted, others to be swallowed, and some few to be
`chewed and digested: that is, some books are to be read only in parts; oth-
`ers to be read but not curiously; and some few to be read wholly, and with
`diligence and attention.
`
`I would like to think that this book might fit any of the above descriptions, depending on the
`reader’s need.
`Finally, I would like to thank my wife Alison for her love, understanding and support over
`the time I have spent preparing this book. Now that the book is finished, I should have more
`time to spend with her and my three children, Laura, Joanna and David.
`
`Mark Gibson
`May 2001
`
`IPR2018-00126
`
`Page 9 of 610
`
`I-MAK 1020
`
`

`

`1408_FM 6/10/03 10:46 AM Page ix
`
`Contributors
`
`MARK GIBSON
`
`Mark Gibson, BPharm, PhD, CChem, MRSC, MRPharmS, is currently responsible for par-
`enteral and oral solid dosage form development at AstraZeneca R&D Charnwood, which is a
`division of AstraZeneca. His experience includes formulation and preformulation develop-
`ment, both as a bench scientist and a manger, at Cyanamid (Lederle) for six years, Fisons Par-
`maceuticals for six years, Astra Pharmaceuticals for four years and AstraZeneca for two years.
`He has worked on a variety of dosage forms and routes of delivery, including inhalation, oral,
`nasal, ophthalmic, parenteral and transdermal, resulting in some patents or marketable prod-
`ucts. Dr. Gibson is a member of the UK Academy of Pharmaceutical Sciences, the Parenteral
`Society and the Aerosol Society.
`
`BERTIL AB RAHAMSSON
`
`Bertil Abrahamsson, PhD, leads pharmaceutical and analytical research and development at
`AstraZeneca R&D Mölndal. His experience is in in vitro/in vivo correlations, in vitro dissolu-
`tion, in vivo imaging of dosage forms and drug absorption. Dr. Abrahamsson has published
`more than 20 articles in clinical pharmacology.
`
`KEITH R. BRAIN
`
`Keith Brain, BPharm, PhD, is head of the drug delivery research group at Cardiff Univer-
`sity. His major responsibilities are in molecular recognition research and skin research. He
`is also CEO of An-eX, a university-based spinoff company formed in 1988, which
`specialises in confidential contract research and development for international clients. He
`has organized several international conferences on skin research and workshops on molec-
`ular imprinting.
`
`ix
`
`IPR2018-00126
`
`Page 10 of 610
`
`I-MAK 1020
`
`

`

`1408_FM 6/10/03 10:46 AM Page x
`
`x
`
`Pharmaceutical Preformulation and Formulation
`
`JOANNE BROADHEAD
`
`Joanne Broadhead, PhD, MRPharmS, manages a small team of formulation scientists at As-
`traZeneca R&D Charnwood in the development of solid and parenteral products. Her expe-
`rience includes two years as a formulation scientist for creative BioMolecules (Hopkinton,
`Mass., USA) prior to joining AstraZeneca in 1996. Dr. Broadhead is a member of the Ameri-
`can Association of Pharmaceutical Scientists, the Parenteral Drug Association and the Par-
`enteral Society.
`
`PETER DAVIES
`
`Peter Davies, BPharm, MRPharmS, PhD, is currently the formulation team leader in drug de-
`livery sciences at Roche Discovery Welwyn. At Roche for 18 years, his principal areas of in-
`terest are preformulation and formulation of solid dosage forms.
`
`NIGEL DAY
`
`Nigel H. Day, BSc, MSc, PhD, is a team manager in product development at AstraZeneca R&D
`Charnwood (formerly Fisons R&D and Astra Charnwood). He has wide experience in the de-
`velopment of solid, liquid and inhalation formulations. His professional experience includes
`a postdoctoral research fellowship at the University of Bradford and seven years in the Hoff-
`man-La Roche UK R&D Laboratories. Dr. Day is a member of the UK Academy of Pharma-
`ceutical Sciences.
`
`GERRY STEELE
`
`Gerry Steele, BSc, MSc, PhD, CChem, MRSC, is a team manager at AstraZeneca R&D Charn-
`wood. His research experience includes preformulation work and characterization and the
`surface rheology of phospholipid monolayers using photon correlation spectroscopy. Prior to
`joining AstraZeneca R&D Charnwood, he spent seven years with Fisons plc and a year at In-
`versesk Research International. Dr. Steele is a member of the Royal Society of Chemistry and
`the Academy of Pharmaceutical Scientists (of the Royal Pharmaceutical Society of Great
`Britain).
`
`ANNA-LENA UNGELL
`
`Anna-Lena Ungell, PhD, is associate director of DMPK and bioanalytical chemistry at
`AstraZeneca R&D Mölndal. She supports preclinical and pharmaceutical projects relating to
`absorption as well as the gastrointestinal absorption of drugs. Her experience includes ab-
`sorption models, mechanisms of drug absorption from the intestinal tract, substance evalua-
`tion and basic formulation development. Dr. Ungell has four patents and has written
`numerous research articles and reviews.
`
`IPR2018-00126
`
`Page 11 of 610
`
`I-MAK 1020
`
`

`

`1408_FM 6/10/03 10:46 AM Page xi
`
`Contributors
`
`xi
`
`KENNETH A. WALTERS
`
`Kenneth A. Walters, PhD, is director of An-eX Analytical Services Ltd and an honorary lec-
`turer in pharmaceutical chemistry at the Welsh School of Pharmacy. His research interests are
`biological membrane penetration enhancement and retardation, particularly with respect to
`skin. He has experience at Fisons Pharmaceuticals, Eastman Pharmaceuticals (a division of
`Eastman Kodak Company) and Controlled Therapeutics Ltd. (Scotland). Dr. Walters has
`published many articles and reviews and has co-edited two volumes on skin penetration en-
`hancement and dermal toxicity. He is a charter member of the American Association of Phar-
`maceutical Scientists and also a member of the Society of Investigative Dermatology, the
`Controlled Release Society, the Society of Toxicology and the Society of Cosmetic Scientists.
`
`PAUL WRIGHT
`
`Paul Wright is an associate director within product development at AstraZeneca. He has in-
`halation development responsibilities at both the Charnwood (UK) site and the Lund (Swe-
`den) site. Mr. Wright has extensive experience in major respiratory dosage forms and is a
`committee member of several industry groups.
`
`IPR2018-00126
`
`Page 12 of 610
`
`I-MAK 1020
`
`

`

`IPR2018-00126
`
`Page 13 of 610
`
`I-MAK 1020
`
`IPR2018-00126
`
`Page 13 of 610
`
`I-MAK 1020
`
`

`

`1408_Chapter 01I 6/10/03 10:15 AM Page 1
`
`1
`
`Introduction and Perspective
`
`Mark Gibson
`AstraZeneca R&D Charnwood
`Loughborough, United Kingdom
`
`This book is intended to be a practical guide to pharmaceutical preformulation and formula-
`tion. It can be used as a reference source and a guidance tool for those working in the
`pharmaceutical industry or related industries, for example, medical devices and biophar-
`maceuticals, or anyone wanting an insight into this subject area. The information presented is
`essentially based on the extensive experiences of the editor and various other contributors
`who are all actively working in the industry and have learned “best practice” from their expe-
`riences.
`There are various excellent books already available which cover the theoretical aspects of
`different types of pharmaceutical dosage forms and processes. A variety of books are also
`available that focus on the drug development process, business, regulatory and project man-
`agement aspects. In my opinion, there has been a long-standing need for a pragmatic guide to
`pharmaceutical preformulation and formulation with an emphasis on what practical studies
`need to be undertaken, for what reasons and during what key stages of the drug development
`process. The important stages where preformulation, biopharmaceutics and formulation play
`a key role are candidate drug selection through the various stages of product development.
`This book has been written to try and address this need.
`A logical approach to product development is described in the book, with the key stages
`identified and the preformulation, biopharmaceutics and formulation activities and typical is-
`sues at each stage discussed. Wherever possible, the book is illustrated with real or worked
`
`1
`
`IPR2018-00126
`
`Page 14 of 610
`
`I-MAK 1020
`
`

`

`1408_Chapter 01I 6/10/03 10:15 AM Page 2
`
`2
`
`Pharmaceutical Preformulation and Formulation
`
`examples from contributors who have considerable relevant experience of preformulation,
`biopharmaceutics and formulation development.
`Jim Wells’ book on preformulation (Wells 1988) made a strong impact on trainees and
`pharmaceutical scientists (including myself) working in this field of the pharmaceutical in-
`dustry when it was introduced over 10 years ago. It describes the important concepts and
`methods used in preformulation with the underlying theory. To his credit, Wells’ book is still
`useful today, but sadly, the book is now out of print, and existing copies are hard to obtain. It
`also requires updating to include modern preformulation instrumental techniques which have
`emerged over the last decade, such as thermo gravimetric analysis (TGA), hot stage mi-
`croscopy (HSM), X-ray powder diffraction (XRPD), raman and infra-red spectroscopy and
`solid-state nuclear magnetic resonance (NMR), to name a few. These techniques can be used
`to provide valuable information to characterise the drug substance and aid formulation de-
`velopment using the minimal amounts of compound.
`Pharmaceutical Preformulation and Formulation: A Practical Guide from Candidate Drug
`Selection to Commercial Formulation covers a wider subject area than just preformulation.
`Topics include biopharmaceutics, drug delivery, formulation and process development aspects
`of product development. The book also describes a logical and structured approach to the
`product development process, recommending at what stages appropriate preformulation, bio-
`pharmaceutics and formulation work is best undertaken.
`
`DRUG DEVELOPMENT DRIVERS,
`CHALLENGES, RISKS AND REWARDS
`
`It is important that the reader is aware of the nature of pharmaceutical research and develop-
`ment (R&D) in order to appreciate the importance of preformulation and formulation in the
`overall process.
`In simple terms, the objective of pharmaceutical R&D can be defined as “converting ideas
`into candidate drugs for development”, and the objective of product development defined as
`“converting candidate drugs into products for registration and sale”. In reality, these goals are
`extremely challenging and difficult to achieve because of the many significant hurdles a phar-
`maceutical company has to overcome during the course of drug development. Some of the
`major hurdles are listed in Table 1.1.
`The high risk of failure in drug discovery and development throughout the pharmaceu-
`tical industry statistically shows that, on average, only 1 in 5,000 to 1 in 10,000 compounds
`screened in research will reach the market (Tucker 1984). Of those that are nominated for de-
`velopment, the failure rate will vary from 1 in 5 to 1 in 10 compounds that will achieve regis-
`tration and reach the market-place. On top of that, there is a significant commercial risk from
`those that are marketed; only 3 out of 10 are likely to achieve a fair return on investment. The
`products which give poor return on investment are often the result of poor candidate drug se-
`lection (the compound does not have the desired properties of safety, selectivity, efficacy, po-
`tency or duration) and/or poor product development (the development programme does not
`establish the value of the product). The latter scenario should, and can be, avoided by careful
`assessment at the “product design” stage of development. Product design is discussed further
`in Chapter 5.
`
`IPR2018-00126
`
`Page 15 of 610
`
`I-MAK 1020
`
`

`

`1408_Chapter 01I 6/10/03 10:15 AM Page 3
`
`Introduction and Perspective
`
`3
`
`Table 1.1
`Major hurdles to successful product registration and sale.
`Requirements
`
`Activity
`
`Research
`
`Safety
`
`Clinical
`
`Drug process
`Pharmaceutical
`
`Regulatory
`Manufacturing
`
`Marketing/commercial
`
`Novel compound (patentable?)
`Novel biological mechanism (patentable?)
`Unmet medical needs
`Potent and selective
`High margin of safety
`Non-toxic (not carcinogenic, tetratogenic, mutagenic, etc.)
`Tolerable side-effects profile
`Efficacious
`Acceptable duration of action
`Bulk drug can be synthesised/scaled up
`Acceptable formulation/pack (meets customer needs)
`Drug delivery/product performance acceptable
`Stable/acceptable shelf-life
`Clinical trial process robust and can be scaled up
`Quality of data/documentation
`Manufacturable
`Able to pass pre-approval inspection
`Competitive
`Meets customer needs
`Value for money
`Commercial return
`
`To be successful and competitive, research-based pharmaceutical companies must ensure
`that new discoveries are frequently brought to the market to generate cash flow. This is re-
`quired to fund the next generation of compounds to meet the therapeutic needs of patients,
`and of course, to benefit the shareholders. This cycle of events is sometimes referred to as the
`“product life cycle” and is further illustrated in Figure 1.1.
`The costs of drug discovery and development to bring a New Chemical Entity (NCE) to
`the market are ever increasing. It is currently estimated that in excess of U.S. $500 million is
`required to recoup the costs of research, development, manufacturing, distribution, market-
`ing and sales. A significant proportion of this total is for the cost of failures, or in other words,
`the elimination of unsuccessful compounds. R&D expenditure for an NCE tends to increase
`substantially as the compound progresses from drug discovery research through the various
`clinical trial phases of development. The pivotal Phase III patient trials are usually the largest,
`involving thousands of patients, and hence the most expensive. To reduce development costs,
`
`IPR2018-00126
`
`Page 16 of 610
`
`I-MAK 1020
`
`

`

`1408_Chapter 01I 6/10/03 10:15 AM Page 4
`
`4
`
`Pharmaceutical Preformulation and Formulation
`
`Figure 1.1 Product life cycle.
`
`EXPLORATORY
`RESEARCH
`
`CANDIDATE
`SELECTION
`
`Candidate drug
`selected
`
`STRATEGIC
`RESEARCH
`
`Based on company
`strategy
`
`£/$
`Sales &
`profits
`
`Further market/medical needs
`new indications
`
`Further market/
`medical needs
`product line
`extensions
`
`MARKETING &
`COMMERCIAL
`
`Launch
`
`Regulatory
`submissions
`
`FULL
`DEVELOPMENT
`
`Safety and efficacy
`demonstrated
`
`
`
`
`EXPLORATORYEXPLORATORYEXPLORATORYEXPLORATORY
`
`
`
`DEVELOPMENTDEVELOPMENTDEVELOPMENTDEVELOPMENT
`
`Proof of concept
`demonstrated
`
`some companies selectively screen and eliminate compounds earlier in the drug development
`process based on results from small-scale, less expensive studies in man and progress fewer,
`more certain compounds to later clinical phases.
`In spite of the high risks and high costs involved, there is still a huge incentive for phar-
`maceutical companies to seek the financial rewards from successful marketed products, and
`especially from the phenomenal success of the rare “blockbuster” (reaching sales of ⬎1 billion
`U.S.$ per year). This can earn the company significant profits to reinvest in research and fund
`the product development pipeline.
`Another factor, the risk of delay to registration and launch, can also have a significant im-
`pact on the financial success of a marketed product. McKinsey & Company, a management
`consultancy, assessed that a product that is 6 months late to market will miss out on one-third
`of the potential profit over the product’s lifetime. In comparison, they found that a develop-
`ment cost overspend of 50 percent would reduce profits by just 3.5 percent, and a 9 percent
`overspend in production costs reduced profits by 22 percent (McKinsey & Co. 1991). The loss
`of product revenue is often due to competitor companies being first to market, capturing the
`market share and dictating the market price, in addition to the loss of effective patent life.
`Hence, the importance of accelerating and optimising drug discovery and development, and
`getting to the market first with a new therapeutic class of medicinal product, cannot be un-
`derestimated. The second product to market in the same class will usually be compared with
`the market leader, often unfavourably.
`
`IPR2018-00126
`
`Page 17 of 610
`
`I-MAK 1020
`
`

`

`1408_Chapter 01I 6/10/03 10:15 AM Page 5
`
`Introduction and Perspective
`
`5
`
`The average time from drug discovery to product launch is currently estimated to take
`10 to 12 years. Several factors may have contributed to lengthening development times over
`the years, including an increase in the preclinical phase to select the candidate drug, and also
`an increase in the duration of the clinical and regulatory period required for marketing ap-
`proval. Benchmarking studies show wide gaps between industry average or worst performance
`compared to what is achievable as best practice performance (Spence 1997). On average, the
`preclinical phase currently takes 4 to 6 years to complete, whereas the time from candidate
`drug nomination to regulatory submission takes on average 6 to 8 years, longer for treatments
`of chronic conditions. Most forward-looking pharmaceutical companies are aiming to reduce
`these times by re-evaluation and subsequently streamlining the development process, for ex-
`ample, by introducing more effective clinical programmes and more efficient data reporting
`systems, forward planning and conducting multiple activities in parallel. However, this, in
`turn, may put formulation development and clinical supplies on the critical path, with pres-
`sures to complete these activities in condensed time scales. Suggestions are offered through-
`out this book on how preformulation, biopharmaceutics and formulation can be conducted
`in the most efficient way to avoid delays in development times.
`Any reduction in the total time-frame of drug discovery to market should improve the
`company’s profitability. In a highly competitive market, product lifetimes are being eroded
`due to the pace of introduction of competitor products, the rapid introduction of generic
`products when patents expire and moves to “over-the-counter” (OTC) status. Successful phar-
`maceutical companies are focusing on strategies for optimum “product life cycle manage-
`ment” to maximise the early growth of the product on the market, sustain peak sales for as
`long as the product is in patent and delay the post-patent expiry decline for as long as possi-
`ble. This should maximise the return on investment during a product life cycle to enable the
`company to recover development costs and make further investments in R&D. Figure 1.2
`shows a classic cash flow profile for a new drug product developed and marketed. During de-
`velopment there is a negative cash flow, and it may be some time after launch before sales rev-
`enue crosses from loss to profit because of manufacturing, distribution and advertising costs.
`Profits continue to increase as the market is established to reach peak sales, after which sales
`decrease, especially after the primary patent expires and generic competition is introduced.
`Throughout the life span of a product, it is in a company’s interest to ensure the best
`patent protection in order to achieve the longest possible market exclusivity. Prior to the pri-
`mary patent expiring (normally for the chemical drug substance), it is imperative to introduce
`new indications, formulations, manufacturing processes, devices and general technology,
`which are patent protected, to extend the life of the product and maintain revenue. A patent
`generally has a term of about 20 years, but as development times are getting longer, there will
`be a limited duration of protection remaining once the product is marketed (the effective
`patent life). A comparison of effective patent life for pharmaceutical new chemical entities in
`various countries around the world shows the same downward trend between the 1960s and
`the 1980s (Karia et al. 1992; Lis and Walker 1988).
`Getting to the market quickly is a major business driving force, but this has to be balanced
`with the development of a product of the appropriate quality. There is a need to generate suf-
`ficient information to enable sound decisions on the selection of a candidate drug for devel-
`opment, as well as to develop dosage forms which are “fit for purpose” at the various stages of
`development. Anything more is wasting precious resources (people and drug substance),
`adding unnecessary cost to the programme and, more importantly, extending the development
`time. Perfect quality should not be the target if good quality is sufficient for the intended pur-
`pose. This can only be achieved if there is a clear understanding of the customer requirements.
`
`IPR2018-00126
`
`Page 18 of 610
`
`I-MAK 1020
`
`

`

`1408_Chapter 01I 6/10/03 10:15 AM Page 6
`
`6
`
`Pharmaceutical Preformulation and Formulation
`
`Figure 1.2 Product life cycle management.
`CASH
`FLOW
`
`Primary patent expires
`
`Market
`penetration
`
`Research
`
`Development Launch
`
`Peak
`sales
`
`Increased competitor
`and generic products
`
`Market share held with line
`extensions
`
`No line extensions
`
`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`Increased
`development costs
`
`Manufacturing
`and launch costs
`
`Line extension
`development costs
`
`TIME
`(YEARS)
`
`+
`
`–
`
`For example, if a simple, non-optimised formulation with a relatively short shelf life is accept-
`able for Phase I clinical studies, any further optimisation or stability testing might be consid-
`ered wasteful, unless the data generated can be used later in the development programme.
`There can be a significant risk associated with doing a minimum development pro-
`gramme and cutting corners to fast track to market. Post-launch, the cost of a retrospective fix
`due to poor product/process design and/or development can be extremely high. The addi-
`tional financial cost from work in product/process redevelopment, manufacturing and vali-
`dation, technical support, regulatory and sales and marketing (due to a product recall) can
`easily wipe out the profit from an early launch. This can have several unpleasant knock-on ef-
`fects; it may affect the market share and the company’s relationship with the regulatory au-
`thorities, and its credibility with customers (both externally and internally within the
`company) may be threatened. These factors need to be taken in to account when planning pre-
`formulation/formulation studies which can directly influence the progress of a product to
`market and final product quality.
`
`CURRENT TRENDS IN THE PHARMACEUTICAL INDUSTRY
`
`Increasing competition and threats to the pharmaceutical industry with respect to maintain-
`ing continued sales growth and income mean that successful companies going forward will
`be those which have a portfolio of products capable of showing volume growth. However, to
`show volume growth innovative new products are required. The cost of drug discovery and
`development is escalating because there are no easy targets left and the cost of development
`
`IPR2018-00126
`
`Page 19 of 610
`
`I-MAK 1020
`
`

`

`1408_Chapter 01I 6/10/03 10:15 AM Page 7
`
`Introduction and Perspective
`
`7
`
`and the cost of goods sold is increasing. There have been several mergers and acquisitions of
`research-based pharmaceutical companies,

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