throbber
Remington
`
`Essentials of Pharmaceutics
`
`Edited by Linda A. Felton, PhD
`Chair, Department of Pharmaceutical Sciences
`Associate Professor of Pharmaceutics
`College of Pharmacy
`University of New Mexico
`Albuquerque, NM, USA
`
`Opiant Exhibit 2306
`Nalox-1 Pharmaceuticals, LLC v. Opiant Pharmaceuticals, Inc.
`IPR2019-00685, IPR2019-00688, IPR2019-00694
`Page 1
`
`

`

`Published by Pharmaceutical Press
`1 Lambeth High Street, London SE1 7JN, UK
`
`© Pharmaceutical Press 2012
`
` is a trade mark of Pharmaceutical Press
`
`Pharmaceutical Press is the publishing division of the Royal Pharmaceutical Society
`
`First edition published 2013
`
`Typeset by Newgen KnowledgeWorks, India
`
`Printed in the United States of America by Edwards Brothers Malloy
`
`ISBN 978 0 85711 105 0
`
`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, without the prior written permission of the copyright holder.
`
`The publisher makes no representation, express or implied, with regard to the accuracy of the information
`contained in this book and cannot accept any legal responsibility or liability for any errors or omissions that
`may be made.
`
`A catalogue record for this book is available from the British Library.
`
`This book is adapted from contributions published in Remington: The Science and Practice of Pharmacy 22nd edition.
`
`Opiant Exhibit 2306
`Nalox-1 Pharmaceuticals, LLC v. Opiant Pharmaceuticals, Inc.
`IPR2019-00685, IPR2019-00688, IPR2019-00694
`Page 2
`
`

`

`Chapter 16
`
`
`
` Tonicity, osmoticity, osmolality,
`osmolarity
` Andrew Ingham, MRPharmS, PhD and
`Cathy Y. Poon, PharmD
`
`Chapter 17
`
`
`Chemical kinetics
`Rodney J. Wigent, PhD
`
`277
`
`301
`
`Chapter 18
`
`
`Chapter 19
`
`
`
`Chapter 20
`
`
`Chapter 21
`
`
`317
`Complex formation
` Thorsteinn Loftsson, MSPharm, MSc, PhD
`and Marcus E. Brewster, PhD
`
`Interfacial phenomena
`Paul M. Bummer, PhD and
`Yvonne Perrie, PhD
`
`329
`
`Colloidal dispersions
` Bill J. Bowman, RPh, PhD,
`Clyde M. Ofner III, PhD, Hans Schott, PhD
`and Yvonne Perrie, PhD
`
`343
`
`Coarse dispersions
` James Swarbrick, DSc, PhD,
`Joseph T. Rubino, PhD, RPh and
`Orapin P. Rubino, PhD
`
`Chapter 22
`
`
`Rheology
`Lawrence H. Block, PhD
`
`Chapter 23
`
`
`Powders
` Yi-Bo Wang and
`Robert O. Williams III, PhD
`
`Chapter 24
`
`
`
` Solutions, Emulsions, Suspensions,
`and Extracts
`Michael M. Crowley, PhD
`
`101
`
`Chapter 25
`
`
`
`133
`
` Sterilization Processes and Sterility
`469
`Assurance
` James Agalloco, BEChE, MSChE, MBA,
`William G. Lindboem, Jr., PhD and Russell
`E. Madsen, MS
`
`Chapter 26
`
`
`Parenteral Preparations
`Michael J. Akers, PhD
`
`371
`
`393
`
`411
`
`435
`
`495
`
`533
`
`Contents
`
`Preface v
`About the editor vii
`Contributors ix
`
`1
`
`9
`
`29
`
`37
`
`51
`
`63
`
`81
`
`93
`
` Information resources in pharmacy and
`the pharmaceutical sciences
` Robin H. Bogner, PhD and
`Sharon Giovenale, MSLS
`
`Analysis of medicinals
`Raymond D. Skwierczynski, PhD
`
`Quality assurance and control
` John H. Parker, PhD and
`John E. Enders, PhD, MBA
`
`Stability of pharmaceutical products
` Allan D. Bokser, PhD and
`Patrick B. O’Donnell, PhD
`
`Bioavailability and bioequivalence
`Steven B. Johnson, PharmD
`
`Dissolution
` Vijai Kumar, MS, MBA and
`Praveen Hiremath, PhD
`
` Modern-day drug discovery and
`development
`Rick G. Schnatz, PharmD
`
`The New Drug Approval Process
` Linda A. Felton, PhD and
`Dennis W. Raisch, PhD, RPh
`
` Metrology and pharmaceutical
`calculations
` Roger L. Schnaare, PhD and
`Shelly J. Prince, PhD
`
`Statistics
` Sanford Bolton, PhD and
`Richard Hirsch, PhD
`
`Chapter 1
`
`
`
`Chapter 2
`
`
`Chapter 3
`
`
`Chapter 4
`
`
`Chapter 5
`
`
`Chapter 6
`
`
`Chapter 7
`
`
`
`Chapter 8
`
`
`Chapter 9
`
`
`
`Chapter 10
`
`
`Chapter 11
`
`
`
` Molecular structure, properties and
`states of matter
` Thomas Rades, PhD,
`Keith C. Gordon, PhD and
`Kirsten Graeser, PhD
`
`177
`
`Chapter 27
`
`
`
` Pharmaceutical Compounding – USP
`<797> Sterile Preparations
` Catherine Cone, PharmD, BCPS,
`Linda A. Felton, PhD, Amy Bachyrycz,
`PharmD
`
`Chapter 12
`
`
`Thermodynamics
`Timothy S. Wiedmann, PhD
`
`Chapter 13
`
`
`Solutions and phase equilibria
`Pardeep K. Gupta, PhD
`
`Chapter 14
`
`
`Separation methods
`Loyd V. Allen, Jr., PhD, RPh
`
`207
`
`219
`
`241
`
`Chapter 15
`
`
`Ionic solutions and electrolytic equilibria 257
`Barbara R. Conway, PhD
`
`Chapter 28
`
`
`Ophthalmic Preparations
` Masood Chowhan, PhD, John C. Lang,
`PhD, Paul Missel, PhD
`
`541
`
`Chapter 29
`
`
`Medicated Topicals
`Lawrence H. Block, PhD
`
`Chapter 30
`
`
`Oral Solid Dosage Forms
` Ahmed Adel Sakr, PhD and
`Fars K. Alanazi, PhD
`
`565
`
`581
`
`v
`
`Opiant Exhibit 2306
`Nalox-1 Pharmaceuticals, LLC v. Opiant Pharmaceuticals, Inc.
`IPR2019-00685, IPR2019-00688, IPR2019-00694
`Page 3
`
`

`

`vi
`
`CONTENTS
`
`Chapter 31
`
`
`
`Chapter 32
`
`
`
`Chapter 33
`
`
`Chapter 34
`
`
`Chapter 35
`
`
` Coating of Pharmaceutical Dosage
`Forms
`Stuart C. Porter, PhD
`
` Oral Modified-Release Drug Delivery
`Systems
` Ali R. Rajabi-Siahboomi, PhD,
`Manish S. Rane, PhD and
`Linda A. Felton, PhD
`
`Aerosols
` John J. Sciarra, PhD and
`Christopher J. Sciarra, BS, MS Industrial
`Pharmacy
`
`633
`
`611
`
`Chapter 36
`
`
`Pharmaceutical Excipients
`William J. Reilly, Jr., MBA
`
`Chapter 37
`
`623
`
`
`
` Basic pharmacokinetics and
`pharmacodynamics
` Raymond E. Galinsky, PharmD and
`Craig K. Svensson, PharmD, PhD
`
`Chapter 38
`
`
`Drug action and effect
`Donald N. Franz, PhD
`
`Chapter 39
`
`
`
` Drug absorption, distribution,
`metabolism and excretion
`Michael R. Franklin, PhD
`
`Biotechnology and Drugs
` Ara H. DerMarderosian, PhD and
`Zhiyu Li, PhD
`
`Pharmaceutical Packaging
` C. Jeanne Taborsky, BSChem and
`Kathleen Deiss, RN
`
`651
`
`Chapter 40
`
`
`
`667
`
`Index
`
` Pharmacokinetics/pharmacodynamics
`in drug development
`George L. Drusano, MD
`
`683
`
`705
`
`727
`
`739
`
`755
`
`763
`
`Opiant Exhibit 2306
`Nalox-1 Pharmaceuticals, LLC v. Opiant Pharmaceuticals, Inc.
`IPR2019-00685, IPR2019-00688, IPR2019-00694
`Page 4
`
`

`

`Chapter 4
`Stability of Pharmaceutical Products
`
`Allan D. Bokser, PhD and Patrick B. O’Donnell, PhD
`
`IntroductIon 37
`
`regulatory requIrementS 38
`
`Product StabIlIty 39
`
`PredIctIng Shelf lIfe 45
`
`aPProxImatIonS In aSSeSSIng
`Product StabIlIty 45
`
`PharmaceutIcal contaInerS 47
`
`cloSureS 48
`
`IntroductIon
`
`Stability of a pharmaceutical product may be defined as the
`capability of a particular formulation, in a specific container/
`closure system, to remain within its physical, chemical, mi-
`crobiological, therapeutic, and toxicological specifications at
`a defined storage condition. Pharmaceutical products are ex-
`pected to meet their specifications for identity, purity, quality,
`and strength throughout their defined storage period at specific
`storage conditions. Assurances that the packaged product will
`be stable for its anticipated shelf life must come from an accu-
`mulation of valid data on the drug in its commercial package.
`These stability data include selected parameters that, taken to-
`gether, form the stability profile.
`The stability of a pharmaceutical product is investigated
`throughout the various stages of the development process. The
`stability of a drug substance is first assessed in the preformula-
`tion stage. At this stage, pharmaceutical scientists determine
`the drug substance and its related salts stability/compatibility
`with various solvents, buffered solutions and excipients consid-
`ered for formulation development. Suitable analytical methods
`must be employed in order to ensure the likelihood that this
`assessment will be successful. Optimization of a stable formula-
`tion of a pharmaceutical product is built (using statistical de-
`sign) upon the information obtained from the preformulation
`stage and continues during the formulation development stages.
`Typically, the first formulation development stage may be
`for preclinical studies or as late as the preparation of a “first
`in human” formulation which is often a non-elegant formula-
`tion optimized for short-term dose-ranging clinical studies. The
`second major formulation development stage occurs to support
`Phase II clinical studies (proof of concept phase). The pharma-
`ceutical product developed at this stage is usually the prototype
`for the commercial product. Therefore, the pharmaceutical
`product will be formulated based in part on the stability infor-
`mation obtained from the previous formulations and must meet
`stability requirements for longer-term clinical studies. In the fi-
`nal formulation development state for Phase III clinical studies,
`the formulation must be truly representative of what the com-
`mercial pharmaceutical product will be in order to avoid delays
`in approval. In addition to building on the clinical requirements
`of the drug, the commercial pharmaceutical product must also
`incorporate the commercial or the final market image of the
`product, which includes the container closure system. The sta-
`bility of this product must be demonstrated to the appropriate
`regulatory agencies in order to assign an expiration period and
`date for the product. This expiration period allows for the as-
`signment of an expiration date based on the manufacture date
`of each lot of drug product.
`Once a pharmaceutical product has gained regulatory ap-
`proval and is marketed, the pharmacist must understand the
`proper storage and handling of the drug. In some cases, a phar-
`macist may need to prepare stable compounded preparations
`from this product.
`
`Most drug products are not shipped directly from the man-
`ufacturer to a pharmacy. Typically, a drug product is shipped
`from a manufacturer to a distribution center. From the distri-
`bution center the drug product is then shipped to a wholesaler.
`From the wholesaler, the drug product may be shipped to the
`distribution center for a pharmacy chain or directly to the phar-
`macy. Finally, the drug product is dispensed by the pharmacy
`to the patient. Dispensing of the drug product may be at a hos-
`pital, a clinic, and a traditional “brick and mortar” pharmacy or
`from a mail-order pharmacy. Therefore, the stability typically
`must also assess the robustness of the drug product through its
`supply chain. It is not unusual for temperature excursions to
`occur during these transfers of control.
`Inventory control, or holding, of each drug is important for a
`wholesaler or pharmacy. A drug must be within its expiration
`dating throughout its use by the patient. Solid oral dosages may
`be dispensed in the commercial packaging or in a pharmacy
`supplied container closure system. Most prescriptions are sup-
`plied to patients for up to 30 or 90 days by traditional and mail-
`order pharmacies, respectively. Inventory control of product by
`wholesalers and pharmacies must assess how much dating must
`remain on a product for it to be useful for its customer. This
`causes the actual holding of a product to be shorter than the
`expiration date. Under normal circumstances it is unusual for a
`pharmacy to accept any product with less than 6 month dating
`remaining on a product.
`Much has been written about the development of a stable
`pharmaceutical product. Comprehensive treatments of all as-
`pects of pharmaceutical product stability have been published
`by Connors et al.1, Carstensen2 and more recently by Allen.3
`This chapter will outline the appropriate steps from prefor-
`mulation to drug approval to assure that the pharmaceutical
`product developed is stable. Requirements for compounded
`products will also be discussed.
`The United States Pharmacopeia (USP) General Chapter
`<1191>4 defines the stability of a pharmaceutical product as
`“extent to which a product retains, within specified limits, and
`throughout its period of storage and use (i.e., its shelf life), the
`same properties and characteristics that it possessed at the
`time of its manufacture.” There are five types of stability that
`must be considered for each drug (Table 4-1).
`The use of kinetic and predictive studies for establishing
`credible expiration dating for pharmaceutical products is now
`accepted worldwide. Scientifically designed studies using reli-
`able, meaningful, and specific stability-indicating assays, ap-
`propriate statistical concepts, and a computer to analyze the
`resulting data are used to determine an accurate and realistic
`shelf life. In this way the maximum amount of valid information
`is obtained to establish a reliable, defendable expiration date
`for each formulation. The assigned expiration date is a direct
`application and interpretation of the knowledge gained from
`the stability study.
`Although there are exceptions, 90% of labeled potency
`generally is recognized as the minimum acceptable potency
`
`37
`
`Opiant Exhibit 2306
`Nalox-1 Pharmaceuticals, LLC v. Opiant Pharmaceuticals, Inc.
`IPR2019-00685, IPR2019-00688, IPR2019-00694
`Page 5
`
`

`

`veterinary drugs, are updated periodically in light of current
`knowledge and technology.
`
`comPendIa
`The USP also contains extensive stability and expiration dat-
`ing information. Included are a discussion of stability consid-
`erations in dispensing practices and the responsibilities of both
`the pharmaceutical manufacturer and the dispensing pharma-
`cist. It now is required that product labeling of official articles
`provide recommended storage conditions and an expiration
`date assigned to the specific formulation and package. Official
`storage conditions as defined by the USP 349 are as follows:
`
`• Freezer – a place where temperature is maintained ther-
`mostatically between –25°C and –10°C.
`• Cold – any temperature not exceeding 8°C and refrigera-
`tor is a cold place where the temperature is maintained
`thermostatically between 2 and 8°C.
`• Controlled Cold Temperature – temperature maintained
`thermostatically between 2° and 8°C.
`• Cool is defined as any temperature between 8 and 15°C.
`• Room Temperature – the temperature prevailing in a
`working area.
`• Controlled Room Temperature – temperature maintained
`thermostatically between 20 and 25°C.
`• Warm – any temperature between 30 and 40°C.
`• Excessive Heat – any heat above 40°C.
`• Protect from Freezing – where, in addition to the risk
`of breakage of the container, freezing subjects an article
`to loss of a product to a loss of strength, potency or to
`destructive alteration its characteristics, the container
`label should bear appropriate instructions to protect the
`product from freezing.
`• Dry Place – denotes a place that does not exceed 40%
`average relative humidity (RH) at Controlled Room Tem-
`perature or the equivalent water vapor pressure at other
`temperatures.
`
`Most drug products are stored at controlled room tempera-
`ture and are labeled as such. As is noted above in USP 34, the
`definition of controlled room temperature was a “temperature
`maintained thermostatically between 20 and 25°C (68 and
`77°F).” This definition was established to harmonize with in-
`ternational drug standards efforts. The usual or customary tem-
`perature range is identified as 20 to 25°C, with the possibility of
`encountering excursions in the 15 to 30°C range and with the
`introduction the mean kinetic temperature (MKT).
`The mean kinetic temperature is calculated using the follow-
`ing equation:
`
`
`
` 
`
`−
`
`∆
`/
`H RT
`n
`
`−
`1
`
`e
`
`+
`
`e
`
`−
`
`∆
`/
`H RT
`n
`
`−
`
`∆
`/
`H RT
`1
`
`e
`
`+
`
`−
`
`e
`
` 
`
`In
`
`
`
`Tk
`
`=
`
`(1)
`
`
`
`
`
`∆
`/
`H R
`∆
`/
`+
`+
`H RT
`
`2
`−
`n
`
`
`
`in which Tk is the mean kinetic temperature; ΔH is the heat of ac-
`tivation, 83.144kJ·mole–1; R is the universal gas constant, 8.3144
`× 10 - 3 kJ· mole–1· degree–1; T1 is the value for the temperature
`(in degrees Kelvin (°K)) recorded during the first time period, T2 is
`the value for the temperature recorded during the second time pe-
`riod, e.g., second week; Tn–1 is the value of the second to last time
`period, and Tn is the value for the temperature recorded during
`the nth time period. Typically, the time period is in days or weeks.
`Mean kinetic temperature determine the thermal expose of
`a material. This allows an acceptable estimation to assess if a
`temperature excursion (or series of excursions) adversely af-
`fected a material.
`FDA and ICH Guidelines provide recommendations for
`
`1. the design of stability studies to establish appropriate
`expiry and product storage requirements
`
`38
`
`PHARMACEUTICAL ANALYSIS AND QUALITY CONTROL
`
`Type of Stability
`Chemical
`
`Physical
`
`Microbiological
`
`Table 4-1. types of Stability
`Conditions Maintained Throughout the Shelf Life
`of the Drug Product
`Each active ingredient retains its chemical
`integrity and labeled potency, within the
`specified limits.
`The original physical properties, including
`appearance, palatability, uniformity,
`dissolution, and suspendability are retained.
`Sterility or resistance to microbial growth
`is retained according to the specified
`requirements. Antimicrobial agents that
`are present retain effectiveness within the
`specified limits.
`The therapeutic effect remains unchanged.
`No significant increase in toxicity occurs.
`
`Therapeutic
`Toxicological
`
`level over the shelf life of a drug product. Exceptions to this
`minimum potency include drugs with active pharmaceutical
`ingredients that have a narrow therapeutic thresholds and
`biologics.
`The stability of a commercial pharmaceutical product is
`expressed as an expiration date (expiry). Expiration dating is
`defined, therefore, as the time in which a drug product in a
`specific packaging configuration will remain stable when stored
`under recommended conditions. This date is usually calculat-
`ed by adding the established expiration period to the date of
`manufacture. The date of manufacture is many times defined as
`the date in which the active pharmaceutical ingredient is first
`combined with a drug product excipient.
`An expiration date, which is expressed traditionally in
`terms of month and year, denotes the last day of the month.
`In the United States, the expiration date shall appear on the
`immediate container and the outer retail package. However,
`when single-dose containers are packaged in individual car-
`tons, the expiration date may be placed on the individual
`carton instead of the immediate product container. If a dry
`product is to be reconstituted at the time of dispensing, expi-
`ration dates are assigned to both the dry mixture and the re-
`constituted product. Tamper-resistant packaging is to be used
`where applicable.
`
`regulatory requIrementS
`
`Stability study requirements and expiration dating are covered
`in the Current Good Manufacturing Practices (cGMPs),5 the
`USP,6 and the FDA and ICH guidelines.
`
`good manufacturIng PractIceS
`The GMPs5 as stated in US 21 CFR part 211.166 requires that
`there shall be a written testing program designed to assess the
`stability characteristics of drug products. The results of such
`stability testing shall be used to determine appropriate stor-
`age conditions and expiration dating. The latter is to ensure
`that the pharmaceutical product meets applicable standards of
`identity, strength, quality, and purity at time of use. Regulatory
`agencies of other regions have similar requirements. For ex-
`ample Canadian regulations require that every distributor and
`importer shall monitor, by means of a continuing program, the
`stability of the drug in the package in which it is sold.7 The Eu-
`ropean Union GMPs states “after marketing, the stability of the
`medicinal product should be monitored according to a continu-
`ous appropriate program that will permit the detection of any
`stability issue (e.g., changes in levels of impurities or dissolu-
`tion profile) associated with the formulation in the marketed
`package.”8 These regulations, which apply to both human and
`
`Opiant Exhibit 2306
`Nalox-1 Pharmaceuticals, LLC v. Opiant Pharmaceuticals, Inc.
`IPR2019-00685, IPR2019-00688, IPR2019-00694
`Page 6
`
`

`

`2. the submission of stability information for investigational
`new drugs, biologics, new drug applications, and biologi-
`cal product license applications.
`
`Thus, the guidelines represent a framework for the experi-
`mental design and data analysis as well as the type of docu-
`mentation needed to meet regulatory requirements in the
`drug-development process.
`The International Conference on Harmonization of Technical
`Requirements for Registration of Pharmaceuticals for Human
`Use (ICH) is an ongoing project that brought together regulatory
`authorities and experts from the pharmaceutical industry from
`three regions of the world; Europe, Japan, and the US since its
`inception in 1990. The first conference (ICH1) took place in
`November 1991 in Brussels, and the second conference (ICH2)
`in Orlando, FL, in October 1993. These conferences continue
`to provide an open forum for discussion and resulted in the
`creation of an extensive set of guidelines dealing with the many
`aspects of safety, quality, and efficacy of medicinal products.
`The following ICH Harmonized Tripartite Guidelines related to
`stability are summarized below.
`
`• Q1A (R2) provides the general requirements for Sta-
`bility Testing of New Drug Substances and Products.
`The main thrust of the stability guideline centers on
`criteria for setting up stability protocols, shown in
`Table 4-2. ICH Q1A is considered the parent guideline for
`stability.10
`• Q1B provides the general requirements for assessing the
`intrinsic photostability characteristics of new drug sub-
`stances and drug products.11
`• Q1C provides the reduced general requirements for
`stability testing of new drug products dosage forms that
`use active ingredients already approved in another dosage
`form.12
`• Q1D provides approaches that allows for bracketing and
`matrixing designs for stability testing. This is especially
`important when a new drug product is to be commer-
`cialized with multiple strengths in multiple packaging
`configurations.13
`• Q1E provides acceptable approaches for evaluating stabil-
`ity data to assess the shelf life of the product in a registra-
`tion application.14
`• Q1F was withdrawn. It provided definitions for testing
`conditions to register drugs marketed in Climatic Zones
`III and IV.15 However, countries in Climatic Zone IV
`believed that a larger safety margin was required than
`presented in this guidance.
`
`It should be noted that all ICH guidelines provide approaches
`that are now common in the industry for assessing stability of
`a drug substance or drug product. However, these guidelines
`also make it clear that alternative approaches can be used when
`there are scientifically justifiable reasons.
`
`Table 4-2a. Stability Protocols
`
`Conditions
`Long-term testing 25°C ± 2°C/60%
`RH ± 5%
`Accelerated testing 40°C ± 2°C/75%
`RH ± 6%
`Alternate testinga 30°C ± 2°C/65%
`RH ± 5%
`
`Minimum Time
`Period at Submission
`12 month
`
`6 month
`
`12 month
`
`Required if significant change occurs during 6-mo storage under
`conditions of accelerated testing.
`
`Durations (Months)
`6
`9
`12
`X
`X
`X,
`Y
`O
`
`O
`
`O
`
`0
`R*
`
`1
`
`X
`
`3
`X
`
`O
`
`X
`
`STABILITY Of PHARMACEUTICAL PRODUCTS
`
`39
`
`Table 4-2b. example Stability Pull Schedule for a Solid
`oral dose for Zone I and II
`Storage
`Conditions
`
`18
`X
`
`24
`X
`
`36
`X
`
`25 °C/60
`% RH
`30 °C/65
`% RH
`X,
`40 °C/75%
`Y
`RH
`*from Release testing if testing is within 30 days of stability set down
`R= Release Tests; Appearance (visual); Identity; Assay (HPLC);
`Impurities (HPLC); Dissolution (USP <711>); Moisture Content
`(Karl fischer); Uniformity of Dosage Unit
`X = Tests at Every Stability Pull; Appearance (visual); Assay (HPLC);
`Impurities (HPLC); Dissolution USP <711>
`O = Pull and test only after 40 °C/75% is out of specification;
`Appearance (visual); Assay (HPLC); Impurities (HPLC); Dissolution
`Y = Additional tests periodically performed; Moisture Content (Karl
`fischer)
`
`The FDA and ICH guidelines are readily available on the in-
`ternet. The pharmaceutical scientist can now access a complete
`listing of both FDA and ICH publications. To view the publi-
`cations, go to: http://www.fda.gov/Drugs/GuidanceComplian-
`ceRegulatoryInformation/Guidances/default.htm for the FDA
`guidance webpage, and http://www.ich.org/products/guidelines/
`quality/article/quality-guidelines.html for the ICH guideline
`webpage.
`
`Product StabIlIty
`
`Many factors affect the stability of a pharmaceutical product
`including the intrinsic stability of the active ingredient(s), the
`potential interaction between active and inactive ingredients,
`the manufacturing process, the dosage form, the container-
`liner-closure system and the environmental conditions encoun-
`tered during shipment, storage and handling and length of time
`between manufacture and usage.
`Classically, pharmaceutical product stability evaluations
`have been separated into studies of chemical (including bio-
`chemical) and physical stability of formulations. Realistically,
`there is no absolute division between these two arbitrary divi-
`sions. Physical factors—such as heat, light, and moisture—may
`initiate or accelerate chemical reactions, whereas every time
`a measurement is made on a chemical compound, physical di-
`mensions are included in the study.
`One type of time-related chemical stability failure is a de-
`crease in therapeutic activity of the preparation to below some
`arbitrary labeled content. A second type of chemical stability
`failure is the appearance of a toxic substance, formed as a deg-
`radation product upon storage of the formulation. The numbers
`of published cases reflecting this second type are few. However,
`it is possible, though remote, for both types of stability failures
`to occur simultaneously within the same pharmaceutical prod-
`uct. Thus, the use of stability studies with the resulting applica-
`tion of expiration dating to pharmaceuticals is an attempt to
`predict the approximate time at which the probability of occur-
`rence of a stability failure may reach an intolerable level. This
`estimate is subject to the usual Type 1 or alpha error (setting
`the expiration too early so that the product will be destroyed
`or removed from the market appreciably earlier than actually
`is necessary) and the Type 2 or beta error (setting the date too
`late so that the failure occurs in an unacceptably large pro-
`portion of cases). Thus, it is obligatory that the manufacturer
`clearly and succinctly defines the method for determining the
`
`Opiant Exhibit 2306
`Nalox-1 Pharmaceuticals, LLC v. Opiant Pharmaceuticals, Inc.
`IPR2019-00685, IPR2019-00688, IPR2019-00694
`Page 7
`
`

`

`40
`
`PHARMACEUTICAL ANALYSIS AND QUALITY CONTROL
`
`degree of change in a formulation and the statistical approach
`to be used in making the shelf life prediction. An intrinsic part
`of the statistical methodology must be the statements of value
`for the two types of error. For the safety of the patient a Type 1
`error can be accepted, but not a Type 2 error.
`One type of time related physical stability failures may affect
`the availability or rate of drug release of a product. This type
`of physical stability failure may cause the active ingredient not
`to be released or a higher rate of drug release (dose dumping).
`Another type of time related physical stability failures are ap-
`pearance related. These may just cause the drug product not
`to appear pharmaceutically elegant or may be an artifact of an-
`other physical or chemical stability failure.
`In this treatment, physical and chemical stability are dis-
`cussed along with those dosage form properties that can be
`measured and are useful in predicting shelf life. The effect of
`various physical and chemical phenomena of pharmaceuticals
`also is treated.
`Knowledge of the physical stability of a formulation is very
`important for three primary reasons. First, a pharmaceutical
`product must appear fresh, elegant, and professional, for as long
`as it remains on the shelf. Any changes in physical appearance
`such as color fading or haziness can cause the patient or con-
`sumer to lose confidence in the product. Second, since some
`products are dispensed in multiple-dose containers, potency of
`the active ingredient over time must be ensured for each indi-
`vidual dose. A cloudy solution or a broken emulsion can lead to
`a non-uniform dosage pattern. Third, the active ingredient must
`be bioavailable to the patient throughout the expected shelf life
`of the preparation. A breakdown in the physical system can
`lead to non-availability or “dose dumping” of the medication
`to the patient. In the case of metered-dose inhaler pulmonary
`aerosols, particle aggregation may result in inadequate lung de-
`position of the medication.
`The chemical causes of drug deterioration have been clas-
`sified as incompatibility, oxidation, reduction, hydrolysis,
`racemization, and other mechanisms. In the latter category,
`decarboxylation, deterioration of hydrogen peroxide and
`hypochlorites and the formation of precipitates have been
`included.
`
`PharmaceutIcal doSage formS
`As the various pharmaceutical dosage forms present unique sta-
`bility problems, they are discussed separately in the following
`section.
`tablets
`Stable tablets retain their original size, shape, weight, and color
`under normal handling and storage conditions throughout their
`shelf life. In addition, the in vitro availability of the active ingre-
`dients should not change appreciably with time.
`Excessive powder or solid particles at the bottom of the con-
`tainer, cracks or chips on the face of a tablet, or appearance of
`crystals on the surface of tablets or on container walls are indi-
`cations of physical instability of uncoated tablets. Hence, the ef-
`fect of mild, uniform, and reproducible shaking and tumbling of
`tablets should be studied. The recommended test for such stud-
`ies is the determination of tablet friability as described in the
`USP. Tablet Friability USP <1216> describes the recommended
`apparatus and the test procedure. After visual observation of
`the tablets for chips, cracks, and splits, the intact tablets are
`sorted and weighed to determine the amount of material worn
`away by abrasion. In general, a maximum weight loss of not
`more than 1% of the weight of the tablets being tested is con-
`sidered acceptable for most products. The results of these tests
`are comparative rather than absolute and should be correlated
`with actual stress experience. Packaged tablets also should be
`subjected to cross-country shipping tests as well as to various
`drop tests.
`Tablet hardness (or resistance to crushing or fracturing) can
`be assessed by commercially available hardness testers. As
`
`results will vary with the specific make of the test apparatus
`used, direct comparison of results obtained on different instru-
`ments may not necessarily be made. Thus, the same instrument
`should be used consistently throughout a particular study.
`Color stability of tablets can be followed by an appropriate
`colorimeter or reflectometer with heat, sunlight, and intense ar-
`tificial light employed to accelerate the color deterioration. It is
`still not unusual for color assessment to be performed visually.
`Caution must be used in interpreting the elevated temperature
`data, as the mechanism for degradation at that temperature
`may differ from that at a lower temperature. It is not always
`proper to assume that the same changes will occur at elevated
`temperatures as will be evidenced later at room temperature.
`Cracks, mottling, or tackiness of the coating indicates evidence
`of instability of coated tablets.
`Typically, dissolution is the in vitro test performed to esti-
`mate bioavailability for a tablet regardless of the solubility of
`the active ingredients. Disintegration has been relegated to an
`in-process test or used to help dissolution. Dissolution tests
`should be run in an appropriate medium at 37°C. Actual dis-
`solution conditions, including medium, are developed during
`the clinical development phase of a product. The dissolution
`method developed has to demonstrate a correlation that is
`relevant to the bioavailability of the dosage form. Dissolution
`profiles are examined during development to provide sufficient
`information to define a single sample time point with a mini-
`mum concentration for immediate release product. Controlled
`release drug products require a dissolution profile with concen-
`tration ranges at set sampling points for product assessment.
`When no significant change (such as a change in the polymor-
`phic form of the crystal) has occurred, an unaltered dissolution-
`rate profile of a tablet formulation usually indicates constant in
`vivo bioavailability.
`Uniformity of weight, odor, texture, drug and moisture con-
`tents, and humidity effect may also be studied during a tablet
`stability test.
`gelatin capsules
`Hard gelatin capsules are the type used by pharmaceutical man-
`ufacturers in the production of the majority of their capsule
`products. The pharmacist in the extemporaneous compounding
`of presc

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