throbber

`
`Eye Therapies Exhibit 2027, 1 of 25
`Slayback v. Eye Therapies - IPR2022-00142
`
`

`

`Eye Therapies Exhibit 2027, 2 of 25
`Slayback v. Eye Therapies - IPR2022-00142
`
`

`

`Eye Therapies Exhibit 2027, 3 of 25
`Slayback v. Eye Therapies - IPR2022-00142
`
`

`

`Eye Therapies Exhibit 2027, 4 of 25
`Slayback v. Eye Therapies - IPR2022-00142
`
`

`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`Eye Therapies Exhibit 2027, 5 of 25
`Slayback v. Eye Therapies - IPR2022-00142
`
`

`

`SpecificAtionS 9
`
`coMpendiA 9
`
`MonogrAphS 10
`
`Chapter 2
`Analysis of Medicinals
`
`Raymond D. Skwierczynski, PhD
`
`doSAge forMS—injectAbleS 16
`
`doSAge forMS—orAl drUg prodUctS 17
`
`AdditionAl doSAge forMS 19
`
`releASe And StAbility teSting 10
`
`fUtUre direction 19
`
`UniverSAl teStS 10
`
`Method vAlidAtion for UniverSAl teStS 11
`
`Appendix A 22
`
`Appendix b 24
`
`tAxonoMy 15
`
`The aim of this chapter is to provide the pharmaceutical sci-
`entist with a general overview of the analytical testing that is
`performed in order to release a medicinal product for clinical or
`commercial use and specifications that ensure the quality and
`performance of the medicinal product being released.
`
`SpecificAtionS
`
`The pharmaceutical manufacturer is responsible for ensuring
`the quality, purity, identity, and strength of each lot of drug
`product manufactured. One mandatory control strategy is to
`ensure that the lot “conforms to specification,” which means
`that the drug product (formulated preparation), when tested to
`the listed analytical procedures, will meet the listed acceptance
`criteria.1 Specifications are critical quality standards.
`A specification is a document that is defined as a list of tests,
`references to analytical procedures, and appropriate acceptance
`criteria (numerical range or limit).1 The pharmaceutical manu-
`facturer justifies the information on the specification, which is
`approved by regulatory authorities. Examples of specifications
`for hypothetical drug products are provided in Appendix A.
`Specifications can be categorized in a couple of manners.
`One way is by the intended use of the product. For example,
`is the medicinal an investigational product intended for use
`in a clinical study, or is it a commercial product that will be
`marketed? Let’s consider the evolution of a hypothetical oral
`drug product as it enters the clinic for single- and multiple-
`ascending dose safety studies during Phase 1 clinical studies,
`then multiple-ascending dose efficacy studies in Phase 2a, and
`confirmatory efficacy studies in Phase 2b and Phase 3. In order
`to enter the clinic rapidly, a simple drug product called a drug-
`in-capsule was chosen for Phase 1. This dosage form consists of
`neat drug (no excipients) that has been accurately weighed into
`gelatin capsules. The manufacturing process meets the volume
`demand for Phase 1, but the dosage form must be changed prior
`to scaling-up to Phase 2. Hence, the drug-in-capsule is a “lame
`duck” dosage form. Specifications for investigational products
`tend to have acceptance criteria that reflect the early life of
`the product. Here, the acceptance criteria for dissolution of our
`drug-in-capsule may be “record result.”
`Now let’s say that our drug-in-capsule drug product will be re-
`placed with a dosage form that is manufactured using a scalable
`manufacturing process — a tablet for Phase 2. The tablet will
`first be used in Phase 2 ascending-dose clinical studies, so sev-
`eral dose strengths will need to be manufactured. Let’s say that
`these dose strengths are 10, 25, and 100 mg. The acceptance
`criterion for the dissolution test may remain as “record result”
`because the dosage form is new, or the criterion may be a “di-
`saster check” criterion. Although the analytical chemist may
`
`not be able to assign a strict acceptance criterion because of the
`limited data, a slow-dissolving drug from an immediate-release
`tablet may jeopardize the interpretation of the clinical results.
`So a criterion like 70 percent of the drug dissolved after 60 min-
`utes may be appropriate for the 100-mg tablet. The lower dose
`strengths are usually assigned the same acceptance criterion.
`Finally, the tablet has entered late development and will be
`used for confirmatory studies. The dose ranging Phase 2a stud-
`ies determined that the efficacious dose was 60 mg, so a new 60-
`mg tablet was developed for use in the Phase 3 studies. During
`Phase 3, a robust manufacturing process has been developed at
`suitable scale, a robust dissolution test also has been developed
`and validated, and many lots of tablets have been manufactured
`and analyzed. Now, a suitable acceptance criterion at the time
`of NDA or MAA submission may be 70 percent of the drug dis-
`solved after 30 minutes.
`Another common categorization involves using different ac-
`ceptance criteria for release testing and stability testing in or-
`der to account for changes that may occur during the shelf life.
`For example, a topical cream may become less viscous upon
`storage at 25°C to 30°C for two years. The cream may be too
`fluid to apply when its viscosity is less than 1000 mPa·s. A suit-
`able acceptance criterion for a stability specification may be
`2000 mPa·s. Let’s say that the development data indicate that
`in order to ensure a viscosity of 2000 mPa·s at the end of the
`shelf life, the cream must have a viscosity of 10,000 mPa·s at
`the time of release. Therefore, the acceptance criteria for the
`release and stability specifications would be not less than (NLT)
`10,000 mPa·s and NLT 2000 mPa·s, respectively, in order to en-
`sure a quality product for its entire shelf life.
`
`coMpendiA
`
`All drug products, whether commercial or investigational, must
`meet standards that have been established by Pharmacopeial
`Conventions or Regulatory Agencies. The United States Phar-
`macopeial (USP) Convention is a scientific nonprofit organi-
`zation that sets standards for the quality, purity, identity, and
`strengths of medicines, food ingredients, and dietary supple-
`ments manufactured, distributed, and consumed worldwide.2–4
`USP drug standards are enforceable in the United States by the
`Food and Drug Administration, and these standards are devel-
`oped and relied upon in more than 130 countries.2,4
`The European Pharmacopeia (Ph. Eur.) Commission estab-
`lishes official standards that provide a legal and scientific basis
`for quality control during the development, production, and
`marketing of medicines in 37 signatory states of the conven-
`tion.5,6 In addition to the signatory states, which comprise 26
`countries and the European Union, there are a large number
`
`9
`
`Eye Therapies Exhibit 2027, 6 of 25
`Slayback v. Eye Therapies - IPR2022-00142
`
`

`

`10
`
`PHARMACEUTICAL ANALYSIS AND QUALITY CONTROL
`
`of observers of Ph. Eur. Twenty-two countries (including Aus-
`tralia, Brazil, Canada, China, the Russian Federation, and the
`United States) and the World Health Organization are listed as
`observers. Consequently, the standards developed by Ph. Eur.
`have an impact on the quality of medicines across the globe.5,6
`The USP and Ph. Eur. publish books of pharmacopeial stan-
`dards. The thirty-fourth revision of the USP and the twenty-
`ninth edition of the National Formulary, USP 34-NF29, became
`official on May 1, 2011. The USP-NF is continuously revised.
`Standard revisions are found in supplements to the USP-NF that
`are published twice yearly. Accelerated revisions are published
`in Pharmacopeial Forum (PF) and on the USP website, www.
`usp.org. A new edition of USP-NF becomes official each year
`on May 1. Chapters <1> through <999> are enforceable chap-
`ters, while Chapters <1000> through <1999> are informational
`chapters.3
`The seventh edition of the European Pharmacopeia (Ph. Eur
`7. 0) became official on January 1, 2011. This edition will be
`augmented with eight supplements over a three-year period.5
`The eighth edition of Ph. Eur. will become official on January
`1, 2014. The European Pharmacopeia is published in English
`and French.
`The British Pharmacopeia (BP) is the official collection of
`standards for UK medicinal products and pharmaceutical sub-
`stances.7 The standards are established by the British Pharma-
`copeia Commission. Canada and Australia also use the BP is as
`their official standards. The BP is recognized in over 100 coun-
`tries as an internationally acceptable standard and remains an
`essential reference for all individuals and organizations working
`within pharmaceutical research, development, manufacture,
`and testing across the globe.8 A new edition of the BP becomes
`official each year on January 1.
`Japan’s Ministry of Health, Labor, and Welfare (MHLW) pub-
`lishes The Japanese Pharmacopeia (JP), which provides an of-
`ficial standard to ensure the quality of medicines in Japan.9–11
`The Japanese Pharmacopeia, Sixteenth Edition (JP 16) became
`official on April 1, 2011. JP 16 is printed in Japanese only at the
`time of publication of this chapter. JP 15, however, is available
`in English. Both editions can be found on The Japanese Phar-
`macopeia website and are free of charge.
`
`MonogrAphS
`
`A monograph is written after a drug product has been registered
`and established in the marketplace and usually before its patent
`expires. The USP defines a monograph as a written standard
`that describes an article (e.g., drug substance, drug product,
`excipient, compounded preparation).12 A monograph published
`in any USP compendium provides the name of a substance; its
`definition; package, storage, and labeling requirement; and in-
`formation on tests needed to ensure the substance is of the ap-
`propriate identity, strength, quality, and purity.3 The later part
`of a monograph is similar in scope to a specification.
`The monograph gives manufacturers, governments, and sci-
`entists a public standard by which to judge an article’s quality.
`Monographs play an important role in meeting the USP’s mis-
`sion by providing standards for substances consumed in a glob-
`al marketplace—standards that help maintain public health.
`The USP-NF comprises more than 4000 monographs.12 The
`USP contains monographs for drug substances and preparations
`(drug products); excipient monographs are in the NF.
`The European Pharmacopeia contains more than 2000 gen-
`eral and specific monographs, including chemical substances,
`antibiotics, vaccines, dosage forms, herbal drugs, and homeo-
`pathic preparations.5 One major difference between the Ph.
`Eur. and USP is illustrated by this example: the USP has mono-
`graphs for tablets of a specific drug (e.g., amoxine), whereas Ph.
`Eur. will have only a general monograph for tablets. The BP and
`JP also contain monographs of drug substances and drug prod-
`ucts. The BP, which contains over 3000 monographs, incorpo-
`rates monographs of the European Pharmacopoeia.13
`
`releASe And StAbility teSting
`
`The release and stability testing can be classified in a couple of
`practical ways. One is by the nature of the testing performed:
`chemical, physical, and microbial. For example, the assay for
`the amount of active ingredient in a gel would be a chemical
`test, the viscosity measurement and the in vitro release test
`would be physical tests, and the microbial limits test would be
`a microbiological test. This classification system is often used
`in analytical laboratories that are specialized by the type of test
`being performed. The chapters in pharmacopeia are also orga-
`nized by the nature of the test.3,5,9,10
`Another classification system that is used is based on the at-
`tribute that is being tested. For example, the ICH Guidances and
`the USP provide two categories for the system: analytical tests
`and acceptance criteria for assessing (1) general quality attri-
`butes and (2) product performance.1,14-17 Using the previous gel
`example, the assay, viscosity, and microbial limits tests would
`be examples of tests that assess general quality attributes while
`the in vitro release test would assess product performance.
`
`UniverSAl teStS
`
`There are four tests that are generally applicable to all drug
`products: description, identification, assay, and impurities.
`
`deScription
`This test is often called “appearance” on a specification and is a
`qualitative description of the dosage form. For example, the de-
`scription of a tablet on a specification may read: white, round,
`biconvex, film-coated tablet, imprinted with “400” on one side.
`
`identificAtion
`The purpose of an identification or identity test is to verify the
`identity of the active pharmaceutical ingredient(s) (API) in
`the dosage form. This test should be able to discriminate be-
`tween compounds of closely related structure that are likely to
`be present. Infrared and Raman spectroscopy are commonly
`used techniques. A more practical technique, however, is high-
`performance liquid chromatography because the identity and
`assay can be determined using the same analytical method.
`The ICH Q6A Guidance, however, does not regard identifica-
`tion solely by a single chromatographic retention time as being
`specific, the definition of which is the ability of the method to
`assess unequivocally the analyte in the presence of the sam-
`ple matrix (excipients, impurities, and degradation products).
`Thus, some laboratories will couple a diode array or mass spec-
`trometric (MS) detector to the HPLC system. The diode array
`detector provides a UV spectrum of the drug; the MS detector
`gives the nominal mass of the drug.
`
`ASSAy
`This test determines the strength or content of the API in the
`dosage form and is sometimes called a content test. The method
`should be stability-indicating, which means that the method is
`quantitative and specific and can detect chemical changes with
`respect to time, so that the quantity of the active ingredient(s)
`can be accurately and precisely measured in the presence of
`the sample matrix. HPLC is the most common technique used
`for stability-indicating methods.
`
`iMpUritieS
`ICH Guidance Q6A defines an impurity in a drug product as any
`component that is not the API or an excipient. The most com-
`mon type of impurities that are measured is related substances,
`which are process impurities from the new drug substance syn-
`thesis, degradation products of the API, or both. The test for re-
`lated substances is often referred to as a purity test and must be
`stability indicating. The method can be the same as that used
`for assay, or it can be a different method that has been devel-
`oped for measuring low-level impurities. An additional method
`
`Eye Therapies Exhibit 2027, 7 of 25
`Slayback v. Eye Therapies - IPR2022-00142
`
`

`

`to measure chiral purity may be used to quantify enantiomeric
`impurities in the drug product. In cases in which an organic
`solvent is used during the manufacture of the drug product, a
`method for measuring residual solvents is also used.
`
`Method vAlidAtion for UniverSAl teStS
`
`The objective of validation of an analytical method is to dem-
`onstrate that it is suitable for its intended purpose.3,5,18,19
`Therefore, the objective of the analytical method will govern
`the validation characteristics that need to be evaluated. Typical
`validation characteristics are specificity, accuracy, precision,
`linearity, range, robustness, detection limit, quantitation limit,
`and sample and standard stability. Well-characterized reference
`materials, with documented purity, should be used throughout
`the validation study.19 A validation protocol that describes the
`validation experiments to be performed and their pre-deter-
`mined validation performance criteria is required. In early de-
`velopment, a department Standard Operating Procedure (SOP)
`can be used instead of a protocol, provided that the SOP clearly
`outlines the validation experiments and validation performance
`criteria. Let’s examine the characteristics evaluated for each of
`the four universal tests and typical acceptance criteria that ac-
`company each test on the specification.
`
`ASSAy
`
`Acceptance criteria
`The results from an assay are usually expressed as a percentage
`of the label claim with acceptance criteria that are typically in
`the range from 90.0 to 110.0 percent label claim. For example,
`a manufactured lot of 500-mg aspirin tablets with these accep-
`tance criteria would have an average assay value between 450
`and 550 mg. The width of these limits is to allow for manufac-
`turing variability and shelf-life stability. In some cases, such as
`narrow-therapeutic drugs, the acceptance criteria may be from
`95.0 to 105.0 percent label claim.
`
`validation characteristics
`Assay methods are validated for specificity, linearity, range, ac-
`curacy, precision, robustness, and sample and standard stabil-
`ity.18,19 Validation of detection limit and quantitation limit is
`not needed unless the same method is used to quantify related
`substances.
`
`SPEcificity
`Most assays are performed using HPLC. Specificity can be dem-
`onstrated in a number of ways. A common approach is to spike
`each impurity and excipient into the analytical solution at an
`appropriate level and compare the results of the spiked sam-
`ples against those of the unspiked samples to demonstrate that
`the presence of the impurities or excipients does not affect the
`assay result. Of course, this approach works only if authentic
`samples of the impurities are available.
`If the impurities are not available, the API, or the dosage
`form, is subjected to forced degradation conditions, and the de-
`gradants and synthetic process impurities are separated from
`the API, using the method that is being validated, as well as
`other methods that make use of different stationary phases
`(chromatographic columns) and chromatographic conditions
`(mobile phase components and gradients, flow rate). Diode ar-
`ray or MS detection is often used. The goal here is to demon-
`strate that no impurity co-elutes with the API in the validated
`method.
`The forced degradation conditions that are often used in
`the analytical laboratory for method development and valida-
`tion are dissolving the API (with or without excipients) in an
`aqueous or aqueous-organic solvent and exposing the solution
`to acidic, basic, and nucleophilic and free radical oxidation
`conditions. It is important to control the degradation to less
`than about 10 percent in order to make the forced degradation
`
`ANALYSIS Of MEDICINALS
`
`11
`
`experiments representative of the degradation that might be
`seen during the shelf-life of the drug product and to decrease
`the probability of the degradants reacting with each other or
`degradants reacting with the API.
`In addition, the API and the dosage form should be exposed
`to a variety of moderately stressed stability conditions, such
`as light and prudently selected combinations of heat and hu-
`midity in order to degrade the material “purposefully.” Some
`common storage conditions for purposeful degradation of a sol-
`id-dosage form include 40°C and 75 percent relative humidity
`(40/75) in high-density polyethylene (HDPE) bottles, 40/75 in
`an open dish (containerless), 60/75 in HDPE bottles, and 60/75
`in an open dish. Some pharmaceutical companies have formed
`purposeful-degradation groups that are tasked with identifying
`“soft spots” on API molecules, understanding the degradation
`chemistry, identifying impurities, conducting purposeful degra-
`dation studies, and proposing stabilization strategies to prevent
`or inhibit chemical degradation.20
`If the API is volatile, gas chromatography (GC) can be used
`for the assay test. Just as with HPLC, more than one method
`should be used to demonstrate specificity. Gas chromatography
`with mass spectrometric detection (GC-MS) is useful to accom-
`plish this goal. In cases in which a non-specific assay, such as
`titrimetry or UV spectrophotometry, is used, other supporting
`methods and knowledge of the degradation chemistry can be
`used to demonstrate overall specificity. The use of non-specific
`methods to determine assay, however, is not recommended,
`given the advances in chromatographic and detection technol-
`ogy. The USP is actively modernizing official USP-NF mono-
`graphs for small-molecule API and their dosage forms that use
`outdated analytical technology or have non-specific identifica-
`tion and assay tests.21
`
`LinEarity and rangE
`A linear relationship should be evaluated across the range of
`the analytical method. For a drug product that has assay ac-
`ceptance criteria of 90.0 to 110.0 percent label claim, linearity
`is usually demonstrated minimally from 80.0 to 120.0 percent.
`Some companies have a policy to demonstrate linearity over an
`expanded range from 70.0 to 130.0 percent. The linearity can
`be demonstrated directly on the API or on separate weighings
`of API and excipient mixtures.
`
`accuracy
`The accuracy of an analytical method expresses the closeness
`of agreement between the value found using the method and
`either an accepted reference value or a conventional true value.
`Accuracy is also termed trueness. A common way that accu-
`racy of a method is evaluated is by performing “spike and re-
`covery” experiments. Here, a known quantity of drug substance
`is spiked into a placebo matrix and compared to a reference
`solution that contains a known amount of drug substance. The
`compatibility of the analyte with the filter used during sample
`preparation is often formally evaluated at this time. Another
`common way to validate the accuracy of a method is to com-
`pare the results obtained from the method being validated to
`those obtained from a second well-characterized procedure.
`Accuracy should be assessed using at least nine determi-
`nations over a minimum of three concentration levels (three
`different concentrations with three replicates of each concen-
`tration) that cover the specified range.
`Accuracy can be inferred once precision, linearity, and speci-
`ficity have been established. This approach is more common in
`early development activities, such as IND-enabling toxicology
`studies or Phase 1 clinical studies.
`
`PrEciSion
`The precision of an analytical method expresses the closeness
`of agreement between a series of measurements obtained from
`multiple samplings of the same homogeneous sample under the
`prescribed conditions.18,19 The precision of a method is best
`
`Eye Therapies Exhibit 2027, 8 of 25
`Slayback v. Eye Therapies - IPR2022-00142
`
`

`

`12
`
`PHARMACEUTICAL ANALYSIS AND QUALITY CONTROL
`
`evaluated using authentic homogeneous samples. If authentic
`samples are not available, artificially prepared samples or a
`sample solution may be used.
`Precision is considered at three levels: repeatability, inter-
`mediate precision, and reproducibility, which are intra-assay,
`within-laboratory, and inter-laboratory measurements, respec-
`tively. Repeatability is measured in one of two ways. One is to
`perform a minimum of six determinations at 100 percent of the
`test concentration. The other is to perform at least nine deter-
`minations over a minimum of three concentration levels (three
`different concentrations with three replicates of each concen-
`tration) that cover the specified range. Depending on company
`policy, repeatability and intermediate precision may be the
`only precision evaluation made in early development. Interme-
`diate precision in its simplest sense is a comparison of results
`obtained by two analytical chemists from the same department
`(laboratory), preferably using two different chromatographic
`systems. Intermediate precision, however, can also be a design
`of experiments (DOE) in which variables such as analytical
`chemists, chromatographic systems, and days are examined.
`Reproducibility is usually assessed prior to or during regis-
`tration stability studies. A DOE is often used. Variable studies
`are similar to those used in intermediate precision, except that
`multiple analytical chemists and chromatographic systems are
`from two laboratories. These laboratories are often the labo-
`ratory that developed the method and the laboratory that will
`perform the release testing of the drug product.
`
`robuStnESS
`The robustness of an analytical method is evaluated prior to or
`during registration stability studies. The robustness evaluation
`shows the reliability of the method with respect to deliberate
`changes made to the method. For example, consider an HPLC
`assay method for which the pH of the aqueous buffer used in
`the mobile phase is 4.5. Experiments may be performed using
`buffers at pH 4.0 and 5.0 in order to evaluate the criticality of
`mobile phase pH on the assay results. Other variables that may
`be evaluated in robustness experiments for assay methods are
`flow rate, injection volume, column temperature, sample tray
`temperature, column manufacturer, mobile phase composition,
`extraction time for sample preparation, and solvent (diluents)
`composition for sample preparation. The numbers of variables
`can be large, and the variables may be interdependent, so a
`DOE approach to evaluate robustness is encouraged.
`
`SamPLE and Standard StabiLity
`Regardless of the phase of development, the analytical samples
`and standards need to be stable for the duration of the analy-
`sis. This stability needs to be evaluated formally during method
`validation.
`
`iMpUritieS: relAted SUbStAnceS
`Acceptance criteria
`The acceptance criteria for related substances on a drug prod-
`uct specification are expressed as less than or equal to a numer-
`ical value and are provided for individual impurities and total
`impurities. The purity results are reported as a percentage, with
`respect to either the peak area of the API in the drug product
`or the sum of the integrated peaks on the chromatogram. Pu-
`rity methods must be stability-indicating. Related substances
`arising during drug product manufacturing and from the degra-
`dation of the drug substance must be monitored. Process impu-
`rities originating during the synthesis of the API are normally
`controlled during drug substance testing and are not listed on
`the drug product specification unless the synthesis impurity is
`also a degradation product in the drug product.
`The magnitude of the acceptance criterion and how the im-
`purity is listed on the specification are determined by the total
`daily dose, the dose at which the impurity was qualified in a
`toxicology study, manufacturing capability, and stability of the
`
`drug product.22 If the daily dose is not more than (NMT) 1.0 g,
`impurities greater than 0.1 percent must be reported on the
`Certificate of Analysis (CofA). For daily doses greater than 1.0 g,
`the impurities greater than 0.05 percent must be reported. The
`values for all of the reported impurities will be summed, and this
`sum must meet the acceptance criterion for total impurities.
`Some individual impurities are listed on the specification
`and limited in order to ensure the quality of the drug product.
`These impurities are called specified impurities. If the level of
`a specified impurity exceeds the identification threshold listed
`in ICH Guidance Q3B, then this impurity must be identified us-
`ing a combination of mass spectrometric and nuclear magnetic
`resonance (NMR) spectrophotometric techniques. Such impu-
`rities are called specified identified impurities. If the level of a
`specified impurity does not exceed the identification threshold,
`it does not have to be identified. This specified, unidentified
`impurity is listed on the specification by its relative retention
`time, which is calculated by dividing the retention of the impu-
`rity by the retention time of the API. Identification thresholds
`are determined by the maximum daily dose and the total daily
`intake of the impurity and are provided in Table 2-1. Two ex-
`amples are provided to help interpret the use of this table.
`
`• Example 1: Consider a tablet for which the maximum
`daily dose is 750 μg. One percent of the maximum daily
`dose is 7.5 μg, which is greater than the 5 μg total daily
`intake (TDI) limit. Therefore, any impurity greater than
`5 μg or 0.67 percent must be identified.
`• Example 2: Consider a tablet for which the maximum
`daily dose is 200 μg. One percent of the maximum daily
`dose is 2 μg, which is lower than 5 μg TDI. Therefore, any
`impurity greater than 1.0 percent must be identified.
`
`If a specified, identified impurity exceeds the qualification
`thresholds provided in Table 2-2, the impurity must be quali-
`fied in a toxicology study. The qualification threshold calcula-
`tions from the information in Table 2-2 are analogous to those
`performed for determining identification thresholds. The ac-
`ceptance criterion for an individual impurity should not exceed
`the amount that has been qualified in a toxicology study.
`How are meaningful limits set for each impurity? ICH Guid-
`ance Q6A recommends that the acceptance criteria be based
`on safety and efficacy data at hand at the time of filing. The
`Guidance recognizes that the amount of data batch history data
`at the time of filing is likely insufficient to assess manufacturing
`
`Table 2-1. identification thresholds
`Maximum Daily Dose
`Threshold
`< 1 mg
`1.0% or 5 μg TDI, whichever
`is lower
`0.5% or 20 μg TDI, whichever
`is lower
`0.2% or 2 mg TDI, whichever
`is lower
`0.10%
`
`≥ 1 mg and ≤ 10 mg
`
`> 1 mg and ≤ 2000 mg
`
`> 2000 mg
`
`Table 2-2. Qualification thresholds
`Maximum Daily Dose
`Threshold
`< 10 mg
`1.0% or 50 μg TDI, whichever
`is lower
`0.5% or 200 μg TDI,
`whichever is lower
`0.2% or 3 mg TDI, whichever
`is lower
`0.15%
`
`≥ 10 mg and ≤ 100 mg
`
`> 100 mg and ≤ 2000 mg
`
`> 2000 mg
`
`Eye Therapies Exhibit 2027, 9 of 25
`Slayback v. Eye Therapies - IPR2022-00142
`
`

`

`process capability and recommends that limits do not tightly
`encompass the batch data that are available at the time of filing.
`The acceptance criteria for individual impurities that are
`degradation products are determined by extrapolating the sta-
`bility data that are available at the time of filing and setting the
`limit using a statistical analysis that usually involves calculating
`the upper 95 percent confidence limit.
`The acceptance criterion for total impurities is usually es-
`tablished by taking the acceptance criteria for individual im-
`purities into account. If degradation does not occur in the drug
`product, acceptance criterion for total impurities is set at 2.0 or
`3.0 percent. If degradation does occur, this acceptance criterion
`can be as high as 5.0 or 7.0 percent.
`The Guidance, however, recognizes that the development
`stability data are incomplete at the time of filing and allows for
`the limit to be raised through a qualification toxicology study.
`Likewise, the limit can be lowered to a value that is less than
`the qualified amount if stability data indicate that a lower value
`is justifiable. As more batch history is obtained, process capa-
`bility should be re-evaluated. The Guidance allows for the lim-
`its to be raised or lowered based on sufficient commercial batch
`history.
`
`validation characteristics
`Impurity methods for related substances are validated for
`specificity, linearity, range, accuracy, precision, robustness,
`sample and standard stability, and quantitation limit.18,19 Usu-
`ally detection limit is also a validation characteristic that is also
`determined.
`
`SPEcificity
`The validation of a related substances impurity method is simi-
`lar to that for an assay method, regardless if the method is com-
`bined with the assay method or a stand-alone impurity method.
`The USP is actively modernizing official USP-NF monographs
`for small-molecule API and their dosage forms that do not have
`a stability-indicating impurity method for related substances.
`
`LinEarity and rangE
`A linear relationship should be evaluated across the range of
`the analytical method. For a drug product, linearity is assessed
`from the reporting level of an impurity to 120 percent of the
`specification limit.
`
`accuracy
`Accuracy of an impurity method is evaluated by performing
`“spike and recovery” experiments. If reference material for the
`impurities is not available, it is acceptable in early development
`to spike drug substance into a placebo matrix and compare the
`value obtained to a reference solution that contains a known
`amount of drug substance. The amount of drug substance or
`impurity reference material spiked should be representative of
`the expected range for the im

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