throbber
Advanced Drug Delivery Reviews 59 (2007) 29 – 37
`
`www.elsevier.com/locate/addr
`
`The role of degradant profiling in active pharmaceutical ingredients
`and drug products ☆
`Karen M. Alsante a,⁎, Akemi Ando b, Roland Brown c, Janice Ensing a, Todd D. Hatajik a,
`Wei Kong d, Yoshiko Tsuda b
`
`a Pfizer Global Research and Development, Analytical Research and Development, Eastern Point Road, Groton, CT 06340, USA
`b Pfizer Global Research and Development, Analytical Research and Development, Nagoya, Japan
`c Pfizer Global Research and Development, Analytical Research and Development, Sandwich, Kent, CT13 9NJ, UK
`d Pfizer Global Research and Development, Analytical Research and Development, 700 Chesterfield Parkway West, Q1B, Chesterfield, MO 63017, USA
`
`Received 2 February 2006; accepted 25 October 2006
`Available online 15 November 2006
`
`Abstract
`
`Forced degradation studies are used to facilitate the development of analytical methodology, to gain a better understanding of active
`pharmaceutical ingredient (API) and drug product (DP) stability, and to provide information about degradation pathways and degradation
`products. In order to fulfill development and regulatory needs, this publication provides a roadmap for when and how to perform studies, helpful
`tools in designing rugged scientific studies, and guidance on how to record and communicate results.
`© 2006 Published by Elsevier B.V.
`
`Keywords: Forced degradation; Oxidation; Acid/base hydrolysis; Thermal; Humidity; Stability-indicating; CAMEO
`
`Contents
`
`2.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`1. Regulatory requirements .
`.
`.
`.
`.
`.
`1.1.
`Summary of requirements at the IND stage .
`1.2.
`Summary of requirements for marketing application .
`Forced degradation timing and strategy .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`2.1. Degradation discussion .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3. Degradation prediction tools .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3.1.
`CAMEO .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Experimental approach tools .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`4.1. API .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`4.1.1. Acid .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`4.1.2.
`Base .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`4.1.3. Oxidation .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`4.1.4.
`Thermal/humidity.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`4.1.5.
`Photostability .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`4.2. Drug product
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`4.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`. 30
`. 30
`. 30
`. 30
`. 31
`. 31
`. 31
`. 32
`. 32
`. 32
`. 32
`. 32
`. 33
`. 33
`. 33
`
`☆ This review is part of the Advanced Drug Delivery Reviews theme issue on “Pharmaceutical Impurities: Analytical, Toxicological and Regulatory Perspectives".
`⁎ Corresponding author. Tel.: +1 860 441 5408; fax: +1 860 715 7280.
`E-mail address: karen.m.alsante@pfizer.com (K.M. Alsante).
`
`0169-409X/$ - see front matter © 2006 Published by Elsevier B.V.
`doi:10.1016/j.addr.2006.10.006
`
`Opiant Exhibit 2302
`Nalox-1 Pharmaceuticals, LLC v. Opiant Pharmaceuticals, Inc.
`IPR2019-00685, IPR2019-00688, IPR2019-00694
`Page 1
`
`

`

`30
`
`K.M. Alsante et al. / Advanced Drug Delivery Reviews 59 (2007) 29–37
`
`5.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Stability-indicating method development
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`5.1. Mass balance .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`5.2. Key degradation sample set
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`5.3.
`Stereochemical stability .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`5.4.
`Physiochemical stability .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`5.5.
`Identify degradants .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`6. Degradation database .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Appendix A. General protocol of API and DP degradation experiments .
`Appendix B. Recommended API and DP degradation conditions .
`.
`.
`.
`Appendix C. Relative rate factors of degradation.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`References .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`33
`33
`34
`34
`34
`34
`34
`35
`35
`36
`37
`
`1. Regulatory requirements
`
`From a regulatory perspective, forced degradation studies
`provide data to support the following:
`
`• identification of possible degradants
`• degradation pathways and intrinsic stability of the drug
`molecule
`• validation of stability indicating analytical procedures.
`
`Issues addressed in regulatory guidances include:
`
`• Forced degradation studies are typically carried out using
`one batch of material.
`• Forced degradation conditions are more severe than accelerated
`stability testing such as N50 °C; ≥75% relative humidity; in
`excess of ICH light conditions; high and low pH, oxidation, etc.
`• Photostability should be an integral part of forced degrada-
`tion study design [1].
`• Degradation products that do not form in accelerated or long-
`term stability may not have to be isolated or have their
`structure determined.
`• Mass balance should be considered.
`
`Issues not specifically addressed in regulatory guidance:
`
`• Exact experimental conditions for forced degradation studies
`(temperatures, duration, extent of degradation, etc.) are not
`specified.
`• Experimental design is left to the applicant's discretion.
`
`There are guidances available from the FDA as well as from
`private industry on regulatory requirements for IND and NDA
`filings [2]. This paper gives a global perspective on regulatory
`requirements (e.g., USA, Europe and Japan) based on current
`regulations and guidances.
`
`1.1. Summary of requirements at the IND stage
`
`The reporting of forced degradation study conditions or
`results is not required in Phase 1 or 2 INDs. However,
`preliminary studies are encouraged to facilitate the development
`of stability indicating methodology. It is recommended that
`forced degradation testing outlined in the table in Appendix A
`
`be conducted as early in the development of API and DP as
`possible. Studies can be conducted on the API and develop-
`mental formulations to examine for degradation by thermolysis,
`hydrolysis, oxidation, and photolysis to evaluate the potential
`chemical behavior of the active. A draft guidance document
`suggests that results of one-time forced degradation studies
`should be included in Phase 3 INDs [3].
`
`1.2. Summary of requirements for marketing application
`
`Completed studies of the degradation of the API and DP are
`required at
`the NDA stage,
`including isolation and/or
`characterization of significant degradation products and a full
`written account of the degradation studies performed [4].
`Requirements at the time of registration include [1]:
`
`• Forced degradation products should be accurately charac-
`terized and the reaction kinetics established.
`• Structural elucidation of degradation products should be
`attempted, even if not successful, should be referenced in
`the NDA.
`• Mass balance should be determined or at least attempted.
`• Main band peak purity should be confirmed.
`• Any degradants present in ICH stability samples which are
`greater than the identification threshold should be isolated
`and identified.
`
`Information from these studies should be referenced in the
`filing and should provide:
`
`• degradation pathways of the API, alone and in DP
`• discussion of any possible polymorphic or enantiomeric
`substances, and
`• differentiation between drug related degradation and excip-
`ient interferences.
`
`The tables in Appendices A and B outline general pro-
`tocol of tests and conditions recommended for regulatory
`submissions.
`
`2. Forced degradation timing and strategy
`
`The requirements for forced degradation testing depend on
`project needs and the stage of development of the compound.
`
`Opiant Exhibit 2302
`Nalox-1 Pharmaceuticals, LLC v. Opiant Pharmaceuticals, Inc.
`IPR2019-00685, IPR2019-00688, IPR2019-00694
`Page 2
`
`

`

`K.M. Alsante et al. / Advanced Drug Delivery Reviews 59 (2007) 29–37
`
`31
`
`For example, pre-clinical through phase 2 project needs dictate
`intense method development, and the rate of compound
`attrition is high. Therefore, when developing a rational study
`design, forced degradation deliverables should be focused on
`method development activities, and not isolation and identifi-
`cation of degradants. As a compound progresses into later
`phase 2 through registration, method development activities
`center on optimization. The focus of stress testing should be
`directed to characterization and elucidation of degradants.
`Table 1 and Fig. 1 below outline the timing and strategy for
`carrying out forced degradation experiments.
`
`2.1. Degradation discussion
`
`Degradation background discussion is a critical first step in
`the process. In these initial discussions, degradation predic-
`tion, background knowledge and lessons learned can be
`shared. The purpose of the discussion is to review stability and
`degradation mechanisms for API and DP in a team-based
`environment to be used as a resource tool to aid analysts in
`performing forced degradation studies. Degradation discus-
`
`sions are held to facilitate meeting milestone deliverables,
`such as stability indicating methodology. Participants include
`analysts, process chemists, formulators and discovery repre-
`sentatives. Discussion should be reassessed for API process or
`salt changes, DP formulation changes as well as line extension
`efforts.
`
`3. Degradation prediction tools
`
`3.1. CAMEO
`
`CAMEO [5] is a computer program that predicts the pro-
`ducts of organic reactions given starting materials, reagents
`and conditions (see Fig. 1, Step 1: Predict degradants). The
`analyses cover the following key degradation conditions:
`basic/nucleophilic, acidic/electrophilic,
`radical, oxidative/
`reductive and photochemical as well as mechanistic inter-
`pretations of these reactions. In general, the CAMEO algo-
`rithms have been designed to give product mixtures that err on
`predicting more degradation products than actually observed.
`This is preferable to rules that are too restrictive and reject a
`
`Table 1
`Forced degradation timing
`
`Development timing
`
`Actions
`
`Recommendations/rational study design
`
`Pre-phase 1
`
`Pre-phase 1 + 6–12 weeks or after lab
`experiments are complete
`
`➢ Milestone: Initial IND
`
`Formulation development
`Phase I–Phase II
`
`➢Milestone:
`
`Phase III: ICH stability start
`
`Phase III: ICH stability start
`
`➢ Milestone: registration
`
`Step 1
`• Predict API degradants (Fig. 1, Sec. 1)
`• Design experimental protocol (Fig. 1, Sec. 2)
`• Perform experiments (Fig. 1, Sec. 3)
`
`• Focus on experiments resulting in at least 5–20% degradation
`• If degradation by a certain pathway is not predicted and/or
`experimental data prove it unlikely, minimal effort should be
`exerted on that condition
`
`Step 2
`• Assessment of API forced degradation data
`(Fig. 1, step 4)
`• Selection of key degradants for analytical
`method development (Fig. 1, step 6)
`• Challenge existing analytical methodology
`(Fig. 1, step 4)
`• Update degradation database (Fig. 1, step 8)
`➢ Provide analytical methodology with data to support stability indicating confidence
`➢ Methods supplied with expectation of future method development with change of process and/or salt form
`
`• Forced degradation data used for rugged method development
`• Mass balance not required
`• Track only significant degradants
`• Track and/or ID significant peaks by RRT and MW (LC/MS) only
`• Isolation and structure elucidation at this stage not required
`
`Step 3
`• Comprehensive forced degradation experiments
`for DP and API (Fig. 1, steps 2–3)
`• Review excipient compatibility data
`(Fig. 1, step 2)
`• Challenge existing analytical methodology
`(Fig. 1, step 4–5)
`• Update key degradation sample set for DP and
`API (Fig. 1, step 6)
`• Update degradation database (Fig. 1, step 8)
`➢ Rugged analytical method with high confidence in stability indicating ability, no further method development
`activities expected
`
`• Design experiments to highlight process, salt form and/or
`formulation changes
`• Mass balance and peak purity assessment as necessary for method
`development
`• ID significant peaks by RRT and MW (LC/MS) only unless more work
`is necessary for RRF determination or project needs
`
`Step 4
`• Attempt full characterization of significant
`degradants (Fig. 1, step 7)
`• Update degradation database (Fig. 1, step 8)
`
`• Isolation, mechanistic understanding and structure elucidation
`as required
`• Significant degradants that are fully characterized should include
`those seen on real time stability
`➢ Full characterization of significant degradation products completed
`
`Opiant Exhibit 2302
`Nalox-1 Pharmaceuticals, LLC v. Opiant Pharmaceuticals, Inc.
`IPR2019-00685, IPR2019-00688, IPR2019-00694
`Page 3
`
`

`

`32
`
`K.M. Alsante et al. / Advanced Drug Delivery Reviews 59 (2007) 29–37
`
`mulations change. The tables in Appendices A and B outline
`general protocol of tests and conditions that may be used to gene-
`rate data for regulatory submissions.
`
`4.1. API
`
`The specified stress conditions should result in approximately
`5–20% degradation of the API or represent a reasonable maxi-
`mum condition achievable for the API. The specific conditions
`(intensity and duration) used will depend on the chemical
`characteristics of the API. The stressed sample should be
`compared to the unstressed sample (control) and the appropriate
`blank. A compound may not necessarily degrade under a given
`stress condition. No further stressing is advised in these cases [2].
`
`4.1.1. Acid
`Example acids include HCl or H2SO4 (0.1–1 mol/L
`solution). Studies should be carried out in the solution state.
`For certain APIs that are partially soluble or insoluble in the
`described acidic solution, addition of an appropriate co-solvent,
`or adjustment of solution pH in the acidic range may be required
`to achieve dissolution; or the APIs can be run as suspensions
`[2]. Special attention to the API structure should be paid when
`choosing the appropriate co-solvent (i.e. do not use alcohols for
`acidic conditions due to their reactivity). Dimethylsulfoxide,
`acetic acid and propionic acid are useful under acidic
`conditions. Additionally,
`the sample may be heated for a
`defined time/temperature to accelerate degradation, depending
`on the API sensitivity to heat.
`
`4.1.2. Base
`Example bases include NaOH, LiOH or KOH (0.1–1 mol/L
`solution). Studies should be carried out in the solution state. For
`certain APIs which are partially soluble or insoluble in the
`described basic solution, addition of an appropriate co-solvent, or
`adjustment of solution pH may be required to achieve dissolution;
`or the APIs can be run as suspensions. Glyme and 1, 4-dioxane
`facilitate reactions in basic conditions [7]. Additionally, the sample
`may be heated for a defined time/temperature to accelerate
`degradation, depending on the API sensitivity to heat.
`
`4.1.3. Oxidation
`Oxidation can be carried out under an oxygen atmosphere or
`in the presence of peroxides. The use of oxygen is a more
`realistic model. Free radical initiators may be used to accelerate
`oxidation. Generally, a free radical initiator and peroxide will
`produce all primary oxidation degradation products observed
`on real-time stability. Therefore, free radical and/or hydrogen
`peroxide conditions are strongly recommended at all stages of
`development.
`For solution state stress conditions, dissolve the API utilizing
`an appropriate solvent, add 5–20 mol% of a free radical initiator
`at atmospheric pressure. To increase the solubility of oxygen in
`the solution, the reaction can be performed in a reaction vessel
`pressurized at 50–300 psi with molecular oxygen. Additionally,
`the system is heated to accelerate degradation. The temperature
`depends on the free radical initiator selected.
`
`Fig. 1. Forced degradation process flow map — prediction to documentation in
`a structure searchable global degradation database.
`
`key product observed in actual degradation or ICH stability
`studies. It is also likely that certain products predicted can
`undergo further decomposition. Due to these limitations with
`this prediction program, tracking historical degradation data
`in terms of functional groups along with CAMEO prediction
`data provides a more thorough approach to degradation predic-
`tion exercises.
`
`4. Experimental approach tools
`
`Forced degradation studies of API and DP include appropriate
`solid state and solution state stress conditions (e.g. acid/base
`hydrolysis, heat, oxidation, and light exposure) in accordance
`with ICH guidelines (Fig. 1, Steps 2 and 3: Design protocol and
`perform experiments) [1,6]. Forced degradation studies should be
`conducted whenever a stability indicating method is required.
`Studies may need to be repeated as methods, processes, or for-
`
`Opiant Exhibit 2302
`Nalox-1 Pharmaceuticals, LLC v. Opiant Pharmaceuticals, Inc.
`IPR2019-00685, IPR2019-00688, IPR2019-00694
`Page 4
`
`

`

`K.M. Alsante et al. / Advanced Drug Delivery Reviews 59 (2007) 29–37
`
`33
`
`For peroxide conditions, hydrogen peroxide reagent (up to
`3%) can be used. As previously indicated, the addition of an
`appropriate co-solvent may be necessary, depending on API
`solubility. Hydrogen peroxide stress testing can be useful in DP
`studies where hydrogen peroxide is an impurity in an excipient.
`Solid-state stress conditions may be similarly investigated by
`placing the API (as is) in suitable closed containers filled with an
`oxygen headspace versus an argon or nitrogen control headspace.
`Additionally, the sample may be heated for a defined time/
`temperature to accelerate degradation, depending on the API
`sensitivity to heat.
`For later stage development compounds when more time and
`effort can be focused on mechanistic understanding,
`the
`following oxidation conditions can be applied. The addition
`of metal ions to solutions of API can indicate whether there is a
`tendency for the API to be catalytically oxidized. Iron and
`copper ions are routinely found in APIs and formulation
`excipients [8]. Transition metal ions can also reduce peroxide to
`generate hydroxyl
`radicals in a Fenton-type reaction.
`In
`addition,
`light can also effect oxidation reactions. Light
`absorbed by a photosensitizer can react with molecular oxygen
`to form the more reactive singlet oxygen species.
`
`4.1.4. Thermal/humidity
`Solid state stability can be evaluated utilizing accelerated
`storage temperatures in general greater than 50 °C and N75%
`relative humidity. The duration of exposure is dependent on the
`API sensitivity. If the forced degradation thermal/humidity
`conditions produce a phase change, it is recommended to also
`run thermal/humidity conditions below the critical thermal/
`humidity that produces the phase change.
`Arrhenius kinetics may be used to establish an appropriate
`temperature and maximum duration of thermal degradation
`studies. Using an appropriate assumption of activation energy,
`the duration of controlled room temperature storage that is
`simulated by the study can be estimated. The table in Appendix
`C provides a guide to that conversion. In general, an activation
`energy assumption of 15 kcal/mol is recommended. In certain
`preclinical
`through phase 2 studies, an activation energy
`assumption between the recommended 15 kcal/mol assumption
`and an aggressive assumption of 18 kcal/mol might be
`appropriate. In studies where particular concerns exist, an
`activation energy assumption between the recommended
`15 kcal/mol assumption and a conservative assumption of
`12 kcal/mol might be appropriate. Deviation from Arrhenius
`kinetics is increasingly expected above 70–80 °C, and the
`impact of this should be considered during experimental design.
`
`4.1.5. Photostability
`Perform studies in accordance with ICH photostability
`guidelines [9]. Option 1 and/or Option 2 conditions can be
`used. According to the ICH guideline, “the design of the forced
`degradation experiments is left
`to the applicant's discretion
`although the exposure levels should be justified. The recom-
`mended exposures for confirmatory stability studies are an
`overall illumination of not less than 1.2 million lux hours and an
`integrated near ultraviolet energy of not less than 200 W-h/m2.
`
`For forced degradation studies, the samples should be exposed to
`at least 2× the ICH exposure length to ensure adequate exposure
`of the sample. For solution studies, acetonitrile is the co-solvent
`of choice. Methanol can produce more artifact degradation
`products from methoxy radicals produced from light exposure.
`
`4.2. Drug product
`
`Drug product (DP) degradation cannot be predicted solely
`from the stability studies of the API in the solid state or solution.
`The non-active pharmaceutical ingredients can also react with
`the API or catalyze degradation reactions. Impurities in the
`excipients can also lead to degradation in the DP not originally
`observed in the API. For DP formulations, heat, light, and
`humidity are often used. The DP stress conditions should result
`in approximately 5–20% degradation of the API or represent a
`reasonable maximum condition achievable for a given formu-
`lation. The specific conditions used will depend on the chemical
`characteristics of the DP. For a solid DP, key experiments are
`thermal, humidity, photostability and oxidation, if applicable.
`For solution formulations, key experiments are thermal, acid/
`base hydrolysis, oxidation and photostability. It
`is recom-
`mended to compare stressed samples with unstressed samples
`and an appropriate blank. For DP studies, the blank sample is an
`appropriate placebo. The stressed placebo sample will provide
`information about excipient compatibility.
`It is advised to take kinetic time points along the reaction
`pathway for API and DP degradation studies to determine primary
`degradants and a better understanding of the degradation pathway.
`
`5. Stability-indicating method development
`
`A stability-indicating method is defined as an analytical method
`that accurately quantitates the active ingredients without interfer-
`ence from degradation products, process impurities, excipients, or
`other potential impurities. A method that accurately quantitates
`significant degradants may also be considered stability-indicating.
`A proactive approach to developing a stability indicating HPLC
`method should involve forced degradation at the early stages of
`development with the key degradation samples used in the method
`development process (Fig. 1, Step 4: Challenge methodology).
`Forced degradation should be the first step in method development.
`If forced degradation studies are performed early, method
`development and identification of primary degradation products
`and unknown impurities can be run in parallel. Using this process,
`a validated HPLC analytical assay, mechanisms of degradation,
`and the impurity/degradant
`information for filing can all be
`generated without delays in the project timeline.
`
`5.1. Mass balance
`
`Mass balance is defined in the 1999 ICH Guidelines as
`“adding together the assay value and levels of degradation
`products to see how closely these add up to 100 percent of the
`initial value, with due consideration of the margin of analytical
`error”. Assessment of mass balance may be informative in
`assuring that
`the chosen analytical strategy controls all
`
`Opiant Exhibit 2302
`Nalox-1 Pharmaceuticals, LLC v. Opiant Pharmaceuticals, Inc.
`IPR2019-00685, IPR2019-00688, IPR2019-00694
`Page 5
`
`

`

`34
`
`K.M. Alsante et al. / Advanced Drug Delivery Reviews 59 (2007) 29–37
`
`significant degradants (Fig. 1, Step 5: Evaluate purity/potency).
`The Guidelines recognize that it can be difficult to determine
`mass balance due to unknown analytical precision and
`differences in response factor. Additional guidance on helping
`the analyst obtain or approximate mass balance is given by
`Baertschi et al. [10].
`
`5.2. Key degradation sample set
`
`The key degradation-impurity sample set (Fig. 1, Step 6:
`Select key degradants/track peaks) for a given compound is
`equal to the significant degradants plus process impurities
`which can include intermediates, starting materials, and by-
`products. Process related impurities and known degradation
`products might be available as reference standards for use in
`method development. Unknown degradation products can
`also be critical
`in the development of a stability specific
`method.
`Forced degradation studies may generate complex mixtures
`of degradants, but method development should consider only
`significant degradants. Although project-specific factors may
`influence judgments of degradant significance, Table 2
`describes guidelines that may generally be applied, considering
`the stage of development. A degradant in a degraded sample
`may be judged not significant, and hence discounted, if it fails
`to exceed either threshold described.
`Example 1: A degraded sample from an exploratory API
`forced degradation study shows 25% main band loss. Analysis
`shows a major degradant (16%), a second degradant (3%) and
`several further degradants at levels NMT 2%.
`
`performed to identify the unknown significant degradants, as
`appropriate. The integrity of these analyte peaks can be verified
`by LC/MS selective ion monitoring. The purity of the peaks can
`also be verified by collecting the peak of
`interest and
`chromatographing it by another method (TLC, GC, or a second
`HPLC system). Usually key predictive samples can be gener-
`ated on a large-scale basis for preparative isolation. Small-scale
`synthesis is also an option when a likely structure is identified
`and the chemistry can be accomplished in a few short steps.
`
`5.3. Stereochemical stability
`
`Chiral APIs should be assessed for their stereochemical
`stability during forced degradation studies on a case-by-case
`basis. CAMEO analysis should be reviewed for concerns with
`racemization of chiral centers in the API. If the degradation
`prediction suggests racemization to be likely by any condition,
`stereochemical stability should be explored. APIs with one or
`two chiral centers should be analyzed with a chiral method.
`Based on predictive data and chemical knowledge, choose
`degradation conditions that are most
`likely to convert
`the
`molecule. If the chosen conditions do not invert or racemize the
`API, then chiral analysis does not need to be part of further
`forced degradation protocols. APIs with three or more chiral
`centers most likely convert to diastereoisomers and could be
`analyzed with an achiral method. Stereoisomers should be
`treated like any other API related impurity with respect to quan-
`titation, identification and qualification thresholds, etc. [11].
`
`5.4. Physiochemical stability
`
`• The 16% peak is the largest degradant, representing N60% of
`total degradation is judged significant.
`• Although the 3% component comprises more than 10% of
`total degradation, its level is less than 25% that of the largest
`degradant and is thus judged not-significant.
`• The components at or below 2% are less than 10% of total
`degradation, as well as being less than 25% the level of the
`largest degradant. All are thus judged not significant.
`
`A polymorph appearing in the late stage of drug develop-
`ment may require reformulation, redevelopment of analytical
`method and change of manufacturing procedures. In addition to
`this, some physical form change only occurs in the solid state
`[12]. For these reasons,
`the solid form change should be
`monitored during forced degradation studies. In order to insure
`the solvolysis of the API, solvates and hydrates should be
`stressed in closed and open containers.
`
`Example 2: A forced photodegradation experiment results in
`10% main band loss. Many individual degradants are observed,
`of which the largest is 0.8%.
`
`• Since no individual impurity exceeds 10% total degradation,
`none is judged significant.
`
`Fig. 1 details the process involved in the key degradation-
`impurity sample generation and selection. Work should then be
`
`Table 2
`Significance judgment guidelines for forced degradation studies
`Pre-clinical — Phase 2
`Phase 2 — registration
`
`% of largest degradant
`% of total degradation
`
`API
`
`25%
`10%
`
`Drug product
`
`10%
`10%
`
`API
`
`10%
`10%
`
`Drug product
`
`10%
`10%
`
`5.5. Identify degradants
`
`Degradants structure elucidation is a collaborative effort
`involving the analytical chemist, process chemist and/or
`formulator, as well as the degradation, mass spectrometry and
`NMR experts (Fig. 1, Step 7: Identify degradants) [13].
`Typically, the focus will be on collecting LC/MS data only
`through the Phase 1 clinical stage. At the Phase 2 clinical stage
`and beyond, more time is invested in isolation, synthesis and
`structural
`identity using NMR characterization of
`forced
`degradation products of concern.
`
`6. Degradation database
`
`A structure-searchable degradation profile database has
`been created to compile API/DP degradation data and share
`degradation knowledge globally at Pfizer (Fig. 1, Step 8:
`
`Opiant Exhibit 2302
`Nalox-1 Pharmaceuticals, LLC v. Opiant Pharmaceuticals, Inc.
`IPR2019-00685, IPR2019-00688, IPR2019-00694
`Page 6
`
`

`

`K.M. Alsante et al. / Advanced Drug Delivery Reviews 59 (2007) 29–37
`
`35
`
`Appendix B (continued)
`Acid/base hydrolysis
`API
`Duration
`
`5–20% degradation or 14 days maximum
`
`DP (not necessary if DP is not solution form)
`API
`Formulation dependent (usually at formulation
`concentration
`API concentration)
`+/−2 pH units around the target pH
`pH range
`pH adjustment HCl for low range and NaOH for high range
`Co-solvents
`Not applicable
`(if needed)
`Temperature
`Duration
`
`70 °C
`5–20% degradation or (3 weeks maximum)
`see activation energy chart
`risk level
`to
`compare with correlation to shelf life
`
`Oxidative degradation
`
`API
`⁎Radical
`chain
`initiators
`
`Initiator
`
`API
`concentration
`Initiator
`concentration
`Solvent
`Temperature
`Duration
`
`AIBN (40–60 °C) —
`organic soluble
`initiator; ACVA
`(40–60 °C) or VA-044
`(RT — 40 °C) —
`water soluble initiators
`0.1–20 mg/ml
`
`5–20 mol% of
`API concentration
`Acetonitrile/water
`Ambient — 60 °C
`5–20% degradation or
`14 day maximum
`
`Document degradants and mechanisms) [14]. The database is a
`web-based system. It was developed to address certain key
`goals including: improve communication, minimize duplication
`of effort, and establish a proactive approach to drug stability,
`predict problems and identify trends in data. With the
`degradation profile database, an analyst can search by structure
`and produce tabulated results of all the identified degradants
`observed under each degradation condition. The database also
`contains a substructure search function that allows the analyst to
`retrieve all records on compounds with similar structure. This
`function is extremely powerful
`in that
`it enables better
`prediction of degradation reactions and better use of our library
`of degradation studies previously performed. The following
`data is tracked in the database: compound number, notebook
`reference, salt form, parent structures, degradant structures,
`molecular weights of degradants, degradation experimental
`conditions, HPLC procedure references, conditions for
`performing degradant HPLC screening analysis, structure
`elucidation data, report references, links to electronic report
`files, and proposed mechanisms for degradants.
`
`Appendix A. General protocol of API and DP degradation
`experiments
`
`Condition
`
`API
`
`DP
`
`Solid Solution/
`suspension
`
`Solid (tablets,
`capsules,
`blends)
`
`+
`+
`o
`
`Acid

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