throbber

`
`» Pharmaceutical
`Preformulationand
`Formulation
`
`~_
`
`A Practical Guidefrom Candidate Drug
`
`Selection to Commercial Dosage Form
`
`
`
`CELGENE 2063
`~~ Mark Gibsc
`CELGENE2063
`APOTEX v. CELGENE
`STEX v. CELGENE
`ial
`-— R2023-00512
`IPR2023-00512
`
`
`/.- —
`
`|
`
`

`

`PHARMACEUTICAL
`PREFORMULATION
`AND
`FORMULATION
`
`A Practical Guide from
`Candidate Drug Selection to
`Commercial Dosage Form
`
`Mark Gibson
`
`Editor
`
`IHS® Health Group
`
`Your Enterprise Solution to
`Global Healthcare Knowledge
`
`

`

`director of publications.
`
`[HS® Health Group publishes books focused upon applied technology and regulatory affairs
`impacting healthcare manufacturers worldwide. If you are considering writing or
`contributing to a book applicable to the pharmaceutical, biotechnology, medical device,
`diagnostic, cosmetic, or veterinary medicine manufacturing industries, please contact our
`
`Invitation to Authors
`
`Library of Congress Cataloging-in-Publication Data
`
`Pharmaceutical preformulation and formulation : a practical guide from candidate drug
`selection to commercial dosage form / Mark Gibson,editor.
`p.
`3; cm.
`Includes bibliographic references and index,
`ISBN 1-57491-120-1 (hard ; alk. paper)
`1. Drugs—Dosage forms.I. Gibson, Mark, 1957-
`{DNLM:1. Drug Compounding. 2. Biopharmaceutics—methods.3. Chemistry,
`Pharmaceutical—methods. 4. Dosage Forms. 5. Drug Evaluation. QV 778 P53535 2001]
`RS200 .P425 2001
`615'.14—dce21
`
`Commissioned in Europe by Sue Horwood of Medi-Tech. Publications, Storrington, England, on behalf
`of IHS” Health Group, Denver, Colorado, USA. General Scientific Advisor: Dr. Guy Wingate, UK Quality
`Manager, Computer Systems Compliance, Secondary Manufacturing, Glaxo Wellcome, Barnard Castle,
`
`England.
`
`2001016816
`
`10987654321
`
`ISBN: 1-57491-120-1
`Copyright © 2001 by IHS® Health Group. All rights reserved.
`
`All rights reserved. This book is protected by copyright. Nopart of it may be reproduced,stored in a retrieval
`system,or transmitted in any form or by any means, electronic, mechanical, photocopying, recording, or oth-
`erwise, without written permission from the publisher. Printed in the United States of America.
`Where a product trademark, registration mark, or other protected mark is madein the text, ownership
`of the mark remains with the lawful owner of the mark. No claim,intentional or otherwise, is made by ref-
`erence to any such marksin this book.
`While every effort has been made by IHS® Health Groupto ensure the accuracy ofthe information con-
`tained in this book,this organization accepts no responsibility for errors or omissions.
`
`IHS” Health Group
`15 Inverness Way East
`Englewood, CO 80112-5776, USA
`
`+1-303-662-9101
`Phone:
`+]-303-754-3953
`Fax:
`Orders/on-line catalog:
`www.ihshealthgroup.com
`
`

`

`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
`
`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
`
`15
`
`15
`20
`20
`
`21
`
`22
`
`28
`
`34
`
`4)
`46
`
`48
`49
`
`58
`
`87
`
`88
`
`88
`
`

`

`iv
`
`4.
`
`Pharmaceutical Preformulation and Formulation
`
`Biopharmaceutical Support in Candidate Drug Selection
`Anna-Lena Ungelf 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 F,
`In Vivo Techniques for Studies in Man
`Vehicles for Absorption Studies
`Functional Use of Absorption Models
`References
`
`PARTII: Early Drug Development
`
`5.
`
`Early Drug Development: Product Design
`Mark Gibson
`
`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
`4
`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 II!: From Product Design to Commercial Dosage Form
`
`8.
`
`Product Optimisation
`
`Mark Gibson
`
`Product Optimisation Purpose and Scope
`Excipient and Pack Optimisation Considerations
`
`295
`
`295
`296
`
`

`

`Sources of Information
`
`Expert Systems
`Experimental Design
`Stability Testing
`Developing Specifications
`Process Design, Process Optimisation and Scale-Up
`Validation and Launch
`
`Acknowledgements
`References
`
`Parenteral Dosage Forms
`
`Joanne Broadhead
`
`Guiding Principles for Simple Parenteral Solutions
`Choice of Excipients
`Sterility Cansiderations
`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 fn 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
`
`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
`
`

`

`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
`
`531
`
`

`

`This material may be protected by Copyright law (Title 17 U.S. Code)
`
`

`

`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
`methodsused in preformulation with the underlying theory. To his credit, Wells’ bookisstill
`useful today, but sadly, the book is now outof 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 powderdiffraction (XRPD), raman and infra-red spectroscopy and
`solid-state nuclear magnetic resonance (NMR), to namea 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 developmentaspects
`of product development. The book also describes a logical and structured approach to the
`product development process, recommendingat what stages appropriate preformulation,bio-
`pharmaceutics and formulation workis best undertaken.
`
`DRUG DEVELOPMENT DRIVERS,
`CHALLENGES, RISKS AND REWARDS
`
`It is important that the readeris 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 Re~D can be defined as “converting ideas
`into candidate drugs for development’, and the objective of product development defined as
`“converting candidate drugs into productsfor registration andsale”. In reality, these goals are
`extremely challenging anddifficult to achieve because of the manysignificant hurdles a phar-
`maceutical company has to overcome during the course of drug development. Someof the
`major hurdlesarelisted 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,thefailure rate will vary from 1 in 5 to 1 in 10 compoundsthat will achieve regis-
`tration and reach the market-place. On top ofthat, thereis a significant commercial risk from
`those that are marketed; only 3 out of10 arelikely to achieve a fair return on investment. The
`products which give poor return on investmentare often the result of poor candidate drug se-
`lection (the compound doesnot have the desired properties ofsafety, selectivity, efficacy, po-
`tency or duration) and/or poor product development (the development programmedoes not
`establish the value of the product). The latter scenario should, and can be, avoided bycareful
`assessmentat the “product design” stage of development. Product design is discussed further
`in Chapter5.
`
`

`

`Introduction and Perspective
`
`3
`
`Table 1.1
`Major hurdles to successful product registration and sale.
`
`
`Activity
`Requirements
`
`
`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-approvalinspection
`Competitive
`!
`Meets customer needs
`
`Value for money
`
`Commercial reture
`1ee
`
`To be successful and competitive, research-based pharmaceutical companies must ensure
`that new discoveries are frequently brought to the market to generate cash flow. Thisis re-
`quired to fund the next generation of compounds to meet the therapeutic needsofpatients,
`and of course, to benefit the shareholders. This cycle of events is sometimesreferred to as the
`“productlife cycle” andis further illustrated in Figure 1.1.
`Thecosts of drug discovery and developmentto bring a New Chemical Entity (NCE) to
`the marketare ever increasing. It is currently estimated that in excess of U.S. $500 million is
`required to recoupthecosts of research, development, manufacturing, distribution, market-
`ing andsales. A significant proportionofthis total is for the cost offailures, or in other words,
`the elimination of unsuccessful compounds. R&D expenditure for an NCEtendsto increase
`substantially as the compound progresses from drug discovery research through the various
`clinical trial phases of development. The pivotal PhaseIII patienttrials are usually the largest,
`involving thousands ofpatients, and hence the most expensive. To reduce developmentcosts,
`
`

`

`4
`
`Pharmaceutical Preformulation and Formulation
`
`
`
`Figure 1.1 Productlife cycle.
`
`
` STRATEGIC
`
`Further market/medical needs
`RESEARCH
`
`
`— newindications
` £/$
`
`
`Based on company
`Sales &
`
`
`strategy
`profits
` EXPLORATORY
`
`RESEARCH
`
`
`
`
`MARKETING &
`
`COMMERCIAL
`
`
`
`Further market/
`
`
`medical needs
`Launch
`
`
`— product line
`
`
`
`Regulatory
`CANDIDATE
`extensions
`
`
`submissions
` SELECTION
`
`
`
`FULL
`DEVELOPMENT
`
`
`Candidate drug
`
`selected
`
`
`
`EXPLORATORY
`DEVELOPMENT
`
`
`Safety andefficacy
`demonstrated
`
`Proof of concept
`demonstrated
`
`
`some companiesselectively screen and eliminate compoundsearlier in the drug development
`process based on results from small-scale, less expensive studies in man and progress fewer,
`more certain compoundstolaterclinical phases.
`In spite of the high risks and high costs involved,thereis still a huge incentive for phar-
`maceutical companiesto seek the financial rewards from successful marketed products, and
`especially from the phenomenalsuccessofthe rare “blockbuster”(reachingsales of >1 billion
`U.S.$ per year). This can earn the companysignificant profits to reinvest in research and fund
`the product developmentpipeline.
`Anotherfactor, 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 productthat is 6 monthslate to marketwill 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). Theloss
`of productrevenueis often due to competitor companiesbeing first to market, capturing the
`market share and dictating the market price, in addition to the loss of effective patentlife.
`Hence, the importanceof accelerating and optimising drug discovery and development, and
`getting to the marketfirst with a new therapeutic class of medicinal product, cannot be un-
`derestimated. The second product to market in the sameclass will usually be compared with
`the marketleader, often unfavourably.
`
`

`

`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 phaseto select the candidate drug, and also
`an increase in the duration of theclinical and regulatory period required for marketing ap-
`proval. Benchmarkingstudies show wide gaps between industry average or worst performance
`compared to whatis 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 moreeffective clinical programmes and moreefficient data reporting
`systems, forward planning and conducting multiple activities in parallel. However, this, in
`turn, may put formulation developmentandclinical supplies on the critical path, with pres-
`sures to complete these activities in condensed timescales. Suggestions are offered through-
`out this book on how preformulation, biopharmaceutics and formulation can be conducted
`in the mostefficient way to avoid delays in developmenttimes.
`Any reduction in the total time-frame of drug discovery to market should improve the
`company’s profitability. In a highly competitive market, productlifetimes are being eroded
`due to the pace of introduction of competitor products, the rapid introduction of generic
`products when patents expire and movesto “over-the-counter” (OTC) status. Successful phar-
`maceutical companies are focusing on strategies for optimum “productlife 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
`showsa classic cash flow profile for a new drug product developed ‘and marketed. During de-
`velopmentthereis a negative cash flow, and it may be sometimeafter launch beforesales rev-
`enuecrosses 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.
`Throughoutthe life span of a product, it is in a company’s interest to ensure the best
`patentprotection in order to achieve the longest possible market exclusivity. Prior to the pri-
`marypatent 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 thelife of the product and maintain revenue. A patent
`generally has a term of about20 years, but as developmenttimes are getting longer, there will
`be a limited duration of protection remaining once the product is marketed (the effective
`patentlife). A comparison ofeffective patentlife for pharmaceutical new chemicalentities 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 developmentof 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 whichare “fit for purpose”at the variousstages of
`development. Anything more is wasting precious resources (people and drug substance),
`adding unnecessary cost to the programmeand, more importantly, extending the development
`time. Perfect quality should notbethe target if good quality is sufficient for the intended pur-
`pose. This can only beachievedif there is a clear understanding of the customer requirements.
`
`

`

`6
`
`Pharmaceutical Preformulation and Formulation
`
`SeeEee
`
`Figure 1.2 Productlife cycle management.
`
`CASH
`Primary patent expires
`FLOW
`
`
`
`
`Increased competitor
`Peak
`and generic products
`Teae
`Market
`—_—
`
`
`
`penetration
`
`Development Launch
`(YEARS)
`
`
`15 20)
`Manufacturing
`Line extension
`and launch costs
`development costs
`
`sm Market share held with line
`extensions
`
`Noline extensions
`
`TIME
`
`
`
`
`Research
`
`development cost
`
`
`
`For example,if a simple, non-optimised formulation with a relatively short shelflife is accept-
`able for PhaseI clinical studies, any further optimisationorstability testing might be consid-
`ered wasteful, unless the data generated can beusedlater in the development programme.
`There can be a significant risk associated with doing a minimum development pro-
`grammeandcutting cornersto fast track to market. Post-launch,the costof 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, technica] support, regulatory and sales and marketing (due to a productrecall) 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) maybethreatened. These factors need to be taken in to account whenplanning pre-
`formulation/formulation studies which can directly influence the progress of a product to
`market andfinal product quality.
`
`CURRENT TRENDS IN THE PHARMACEUTICAL INDUSTRY
`
`Increasing competition andthreats to the pharmaceutical industry with respect to maintain-
`ing continued sales growth and income meanthat successful companies going forward will
`be those which havea portfolio of products capable of showing volume growth. However, to
`show volume growth innovative new products are required. The cost of drug discovery and
`developmentis escalating because there are no easy targets left and the cost of development
`
`

`

`Introduction and Perspective
`
`7
`
`and the cost of goodssoldis increasing. There have been several mergers and acquisitions of
`research-based pharmaceutical companies, and increased collaborations and inward licensing
`of products and technologies, in attempts to acquire new leads,to share costs, to reduce the
`time to licence and to maintain growth. Unfortunately, mergers and acquisitionsalso result
`in streamlining and job losses which improveefficiency and decrease overheadcosts at the
`same time.
`There is a changing trend in the nature of the candidate drug emerging from pharma-
`ceutical R&D, from a low molecular weight chemical to a more complex macromolecule (bi-
`ologicals), which can be a peptide, protein, enzyme,antibody, nucleic acid, genetic material or
`a multicomponentvaccine. Someof these compounds have been derived from biotechnolog-
`ical processes to produce biotechnological medicinal products that fight infection and disease.
`The U.S. Food and Drug Administration (FDA) and European Agency for the Evaluation of
`Medicinal Products (EMEA) havealready approved biotechnological medicinal products for
`anaemia,cystic fibrosis, growth deficiency, hepatitis and transplant rejection. Many more are
`being developed to treat cancer, human immunodeficiency virus (HIV) infections and ac-
`quired immunodeficiency syndrome(AIDS), multiple sclerosis and stroke. A majorchallenge
`to the formulator is to develop self-administered formulations to deliver macromolecules
`such as proteins and polypeptides into the body, for example, by the oralor inhalation route.
`More sophisticated drug delivery systems are being developed to overcomethe limita-
`tions of conventional formsof drug delivery systems(e.g., tablets and intravenous[IV] solu-
`tions), to overcome problemsof poor drug absorption, the non-compliance of patients and
`inaccurate targeting of therapeutic agents. One example of an emerging drug delivery tech-
`nology is the use of low-level electrical energy to assist the transport of drugsacross the skin
`in a process known aselectrophoresis. This method could beparticularly useful for the deliv-
`ery of peptides and proteins which are not adequately transported by passive transdermal
`therapy. The drug absorptionrate is very rapid and more controlled compared with passive
`diffusion across the skin. Another example is the improvementin inhaler technology to en-
`sure a moreefficient delivery to the lungs, with minimal drug deposition in the mouth and
`trachea. Theuse ofa breath-actuated aerosol is designed to c#ordinate drug delivery with the
`patient’s inhalation to achieve this. A third example is the use of bioerodable polymers that
`can be implantedor injected within the body to administer drugs from a matrix which can be
`formulated to degrade over a long duration from one day to six months, and do notrequire
`retrieval. Someof these specific delivery systems are explained in moredetail in later chapters
`of this book on the various dosage forms.
`Futuristic drug delivery systems are being developed which hopeto facilitate the trans-
`port of a drug with a carrier to its intended destination in the body and thenreleaseit there.
`Liposomes, monoclonalantibodies and modified viruses are being considered to deliver “Te-
`pair genes” by IV injection to target the respiratory epithelium in the treatmentofcystic fi-
`brosis. These novel drug delivery systems not only offer clear medical benefits to the patient
`but can also be opportunities for commercial exploitation, especially useful if a drug is ap-
`proaching the end ofits patentlife.
`There are pressures on the pharmaceutical industry which affect the way products are
`being developed. For example, there is a trend for more comprehensive documentation to
`demonstrate compliance with current Good Manufacturing Practice (CGMP) and Good Lab-
`oratory Practice (GLP) and to demonstrate that systems and procedures havebeenvalidated.
`The trend is for more information required for a regulatory submission, withlittle flexibility
`for changes once submitted. Therefore, the pressure is for a company to submit early and de-
`velop the product “right first time”.
`
`

`

`8
`
`Pharmaceutical Preformulation and Formulation
`
`In spite of efforts to harmonisetests, standards and pharmacopoeias,thereisstill diver-
`sity between the major global markets—Europe, the United States and Japan—which have to
`be taken in to accountin the design of preformulation and formulation programmes(Anony-
`mous 1993). This is discussed further in Chapter 5 on productdesign.
`Otherpressures facing the pharmaceutical industry are ofa political/economicalor envi-
`ronmental nature. Some governmentsare trying to contain healthcare costs by introducing
`healthcare reforms, which maylead to reducedprices andprofit margins for companies,orre-
`stricted markets where only certain drugs can be prescribed. Although the beneficial effect of
`drugs is not questioned in general, the pressure to contain the healthcare costs is acute.
`Healthcare costs are increasing partly because peopleare living longer and more treatments
`are available. This may influence the commercial price that can be obtained for a new prod-
`uct entering the marketand,in turn, the “cost of goods (CoG)target”. The industry average
`for the CoGtarget is 5 to 10 percent of the commercialprice with pressure to keepit as low as
`possible. This may impact on the choice andcost of raw materials, components and packag-
`ing for the product and the design and cost of manufacturing the drug and product.
`Environmental pressures are to use environmentally friendly materials in products and
`processes and to accomplish the reduction of waste emissions from manufacturing processes.
`A good exampleis the replacementof chlorofluorocarbons (CFCs) propellants in pressurised
`metered dose inhalers (pMDIs) with hydrofluorocarbons (HFAs). The production of CFCs in
`developed countries was banned by the Montreal Protocol (an international treaty) apart
`from “essential uses’, such as propellants in pMDIs, to reduce the damageto the earth’s ozone
`layer. However, there is increasing pressure to phase out CFCsaltogether. Thetransition from
`CFC to HFA products involves a massive reformulation exercise with significant technical
`challenges and costs for pharmaceutical companies involved in developing pMDIs, as de-
`scribed in Chapter 10 “Inhalation Dosage Forms”. However, this can be turned into a com-
`mercial opportunity for some companies which have developed patent-protected delivery
`systemsto extend the productlife cycle of their CFC pMDIproducts.
`
`LESSONS LEARNT AND THE WAY FORWARD
`
`To achieve the best chanceofa fast andefficient development programmeto bring a candi-
`date drug to market, several important messages can be gleaned from projects which have
`gonewell and from companies with consistently good track records.
`There are benefits for pharmaceutical developmentto get involved early with preclinical
`research during the candidate drugselection phase. This is to move away from an “over-the-
`wall” hand-over approachof the candidate drug to be developed from “research”to “develop-
`ment”. The drugselectioncriteria will be primarily based on pharmacological properties such
`as potency,selectivity, duration of action and safety/toxicology assessments. However,if all
`these factors are satisfactory and similar, there may be an important difference between the
`pharmaceutical properties of candidate drugs. A candidate drug with preferred pharmaceuti-
`cal properties, for example, good aqueous solubility, crystalline, nonhygroscopic and good
`stability, should be selected to minimise the challenges involved in developing a suitable for-
`mulation. This is discussed further in Chapter 2.
`Another importantfactoris good long-term planning,ideally from candidate drug nom-
`ination to launch, with consideration for the safety, clinical, pharmaceutical development,
`manufacturing operations and regulatory strategies involved to develop the product. Thereis
`a need for one central, integrated, companyproject plan that has been agreed on byall parties
`
`

`

`Introduction and Perspective
`
`9
`
`with a vested interest in the project. Needless to say, the plan should contain details ofactivi-
`ties, timings, responsibilities, milestones, reviews and decision points. Reviews and decision
`points are requiredat the end ofa distinct activity to ensure thatthe projectis still meetingits
`objectives and should progress to the next stage of development. However, these reviews
`should notcause any delays to the programme,rather, they shouldratify whatis already pro-
`gressing. The traditional sequential phases of product development (see Chapter 2) must be
`overlapped to accelerate the product to market. In reality, plans will inevitably change with
`time; they should be “living” documents which are reviewed and updated at regular intervals
`and then communicatedto all parties. There may be several more detailed, lower-level plans
`focusing on departmentalactivities, e.g., for pharmaceutical development, but these plans
`mustbe linked to the top level central project plan.
`Forward planning should provide the opportunity for a well thought out andefficient ap-
`proachto product development,identifying requirements up frontso as to avoid too much de-
`liberation and backtracking along the way. It also should providea visible communication tool.
`Good planningis supported by adopting a systematic and structured approach to prod-
`uct development. The development process can be broken down into several key defined
`stages—productdesign, process design, product optimisation, process optimisation, scale-up
`and so on. Eachstage will have inputs and outputs as shownin Figure 1.3, a simplified frame-
`workfor product development. The appropriate definition and requirementsat eachstage are
`described in Chapters 5 and8ofthis text.
`As product developmentcan take several years to complete,it is important to havean ef-
`fective document managementsystem in place to record the work. The primary reference
`source for recording experimental work will usually be a laboratory notebook. The work
`should be checked, dated and counter-signed to satisfy GLP and intellectual property re-
`quirements. Experimental protocols are sometimesuseful for defining programmesof work,
`
`
`Figure 1.3 Frameworkfor product development.
`
`>
`PLANNING / DOCUMENTATION
`@ Candidate Drug
`
`i
`
`Product
`
`Product Profile
`Critical Quality Parameters
`
`Peeeticn
`
`ae Quantitative Formula
`* Raw Material / Component Specifications
`
`Process
`
`Process Outline
`———-> Equipment / Facility Definition
`
`-
`=
`Scale-Upfor Clinical Trials
`
`In-Process Controls
`*
`* ProductSpecification
`Scale-Up for Commercial
`Production
`
`Process Validation
`NDA
`@Submission
`
`Manufacture Launch Stock
`
`Phase IV
`
`
`

`

`10
`
`Pharmaceutical Preformulation and Formulation
`
`explaining the rationale for the studies and defining the acceptance criteria. Whenthe studies
`are completed, the results can be reported with reference to the protocol and acceptancecri-
`teria. Laboratory notebooks are referenced in the protocols and reports so that the raw data
`can be retrieved in the event of an audit.
`At the completion of key stages of the work, summaryreports can be written, referencing
`all

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