throbber
CENTER FOR DRUG EVALUATION AND
`
`RESEARCH
`
`APPLICA TION NUMBER:
`

`
`22-350
`
`CHEMISTRY REVIEWg S!
`
`

`

`Onglyza
`(saxagliptin) tablets
`NDA 22-350
`
`Summary Basis for Recommended Action
`
`From Chemistry, Manufacturing, and Controls
`
`Applicant:
`
`Bristol-Myers Squibb Company
`PO. Box 4000
`
`Princeton, NJ 08543-4000
`
`Indication:
`
`Saxagliptin is an orally active reversible dipeptidyl peptidase—IV (DPP—IV)
`inhibitor proposed for the treatment of type 2 diabetes.
`
`Presentation: Onglyza (saxagliptin) tablets are film coated tablets containing 2.5 mg or
`5 mg, .2.»-
`. Both strengths will be available in 30 count (95 ml)
`
`and 500 count (200 m1) white opaque
`bottles with
`desiccant. The 5 mg tablets will be additionally available in aluminum/
`aluminum blisters.
`
`M4)
`
`EER Status: Acceptable, 26-FEB-09
`
`Consults:
`
`Methods Validation — Revalidation by Agency was not requested
`EA — Categorical exclusion granted under 21 CFR §25.3l(C)
`
`Original Submission: 30-JUN-08
`
`Post-Approval Agreements: None
`
`. Background for CMC Section of Application
`
`The CMC portion of this NDA was submitted as part of the ONDQA CMC Pilot
`Program to explore science and risk based approaches to assuring product quality.
`A Comprehensive Quality Overall Summary was provided in Module 2 and an
`expanded pharmaceutical development section was submitted in Module 3,
`Section P2. The applicant applied several Quality by Design (QbD) principles in
`the pharmaceutical development and manufacturing approaches.
`
`Drug Substance:
`
`monohydrate form.
`The drug substance for Onglyza is saxagliptin in
`The formal chemical name is (lS,3S,5S)-2-[(2S)—2—Amino-2-(3-hydroxytricyclo
`[3.3.1 .l3,7]dec-1-yl)acetyl]-2-azabicyclo[3. l .0]hexane—3-carbonitrile,
`monohydrate. The molecular structure is provided as follows:
`
`M4)
`
`
`
`

`

`0 H20
`
`CN
`
`The molecular formula is: C13H25N302 - H20 with a calculated molecular weight
`of 333.43. Saxagliptin has been chemically and structurally characterized using:
`
`-
`
`C.
`C
`
`,,
`
`a
`‘
`
`.3
`D
`
`M4)
`
`"
`
`m4)
`"
`
`”(4}
`
`Saxagliptin drug substance will be manufactured at the BMS facility in Swords,
`Ireland using a -—-—~
`commercial process from / starting materials.
`
`The NDA contained expanded information for the synthesis and process
`development of saxagliptin, using Quality by Design approaches. The approach
`included identification the critical quality attributes of the drug substance,
`identification of critical and “key” process parameters, and the implementation of
`process controls for producing drug substance of the desired quality. Several
`statistically designed experiments (DOES) were utilized in the drug substance
`process development. The application included a design space for drug substance,
`allowing for increased flexibility for manufacturing. Drug substance quality is
`assured through manufacturing process controls combined with conventional end-
`product testing, including appearance, color, identification, and assay and
`impurities/degradants by
`High Performance Liquid
`Chromatography /-HPLC).
`
`.
`
`Based on the 32-month primary stability data for saxagliptin drug substance from
`Process C and the 12-month primary stability data for saxagliptin drug substance
`from Process D, stored at 5°C (i3°C), a If” , period for the drug
`substance is granted with the following label statements:
`
`K
`
`._
`
`.
`
`9 11(4)
`
`Conclusion: Drug substance is satisfactory
`
`

`

`Drug product
`
`Onglyza (saxagliptin) tablets is an immediate release tablet, with the following
`description:
`
`0
`
`0
`
`2.5 mg tablet, containing 2.79 mg saxagliptin hydrochloride (anhydrous):
`round, biconvex yellow tablet printed with “2.5” on one side and “4214”
`on the other side, with a tablet weight of 236mg
`5 mg tablet, containing 5.58 mg saxagliptin hydrochloride (anhydrous):
`round, biconvex pink tablet printed with “5’ on one side and “4215” on the
`other side, with a tablet weight of 239 mg
`
`Based on tablet dimension measurements, saxagliptin film coated tablets, 2.5 mg,
`and saxagliptin film coated tablets, 5 mg, are expected to have an average
`thickness of 4.2 mm and average diameter of 8.2 mm.
`
`11(4)
`
`The saxagliptin film coated tablets contain the following excipients: lactose
`monohydrate, microcrystalline cellulose, croscarmellose sodium, magnesium
`
`state,
`‘
`yellow (2.5 mg), \— pink (5 mg tablet),
`
`,
`No novel excipients are used in the
`manufacture of saxagliptin film coated tablets. The drug loading for both
`strengths is relatively low at less than /
`
`M4)
`
`The drug product is manufactured by a —/‘ coating process in a
`
`
`
`M4)
`
`W)
`
`The enhanced pharmaceutical development provided in the application included
`use of risk assessment and DoEs to evaluate the criticality of process parameters
`and support development of the control strategy and design space. In addition,
`experimental data was leveraged to develop a mechanistic model of the coating
`operation used to predict process performance. Drug product quality is assured
`through manufacturing process controls combined with conventional end-product
`
`

`

`testing, including identification, dissolution, water content, uniformity of dosage
`units, and assay and impurities/degradants by \HPLC.
`
`MM.
`
`Saxagliptin film coated tablets will be packaged and marketed as 30 and 90 count
`in 95 mL and 500 count in 200 mL white, opaque, ’ I’M
`. bottles, with a two piece child resistant closure having an aluminum foil
`induction seal. Each bottle contains a cotton coil and one 2g pouch containing
`silica gel (desiccant) and activated carbon. 5 mg saxagliptin film coated tablets
`will also be packaged and marketed in aluminum/aluminum (alu/alu) blisters.
`
`N4)
`
`Based on provided stability data, the 2.5 mg strength saxagliptin tablets are
`granted a 36 month stability period for 30 and 90 count bottles containing
`desiccant. The 5 mg strength saxagliptin tablets are granted a 36 month stability
`period for 30, 90 and 500 count bottles containing desiccant, and 36 month
`stability period when stored in aluminum/aluminum blisters. Both the 2.5 mg and
`5 mg strengths contain the following labeling statement: “Store at 25°C (77°F);
`excursions permitted to 15°~30°C (59°-86°F) [see USP Controlled Room
`Temperature].
`
`Conclusion: Drug product is satisfactory.
`
`Additional Items:
`
`All associated Drug Master Files are acceptable or the pertinent information has
`been adequately provided in the application.
`
`The analytical methods used in the testing procedures (release, stability and in-
`process) are well known and widely used by the biopharmaceutical
`industry;
`revalidation by Agency laboratories will not be requested.
`
`The application includes design space for both drug product and drug substance.
`
`A comparability protocol for extensive changes that could be made with reduced
`reporting requirements was originally submitted in the application but was later
`withdrawn. The applicant expressed interest in submitting a revised version of the
`comparability protocol as a supplement at a later date following additional
`discussion and clarification from the Agency.
`
`Overall Conclusion: From a CMC perspective, the application is recommended for
`approval.
`.
`
`Christine M. V. Moore, Ph.D.
`Acting Director, DPA I/ONDQA
`
`

`

`This is a representation of an electronic record that was signed electronically and
`this page is the manifestation of the electronic signature.
`
`Christine Moore
`4/17/2009 10:47:05 AM
`CHEMIST
`
`

`

`Overall Summary of Quality
`
`NDA 22-350
`
`Onglyza (saxagliptin) IR tablets
`
`Bristol—Myers Squibb (BMS)
`
`ONDQA CMC Pilot Program Application
`
`CMC Quality Review Team
`
`Sharmista Chatterjee, Ph.D.
`
`John C. Hill, Ph.D.
`
`Prafull K. Shiromani, Ph.D.
`
`Suong T. Tran, Ph.D.
`
`

`

`I. Overview
`
`This Overall Summary of Quality is intended to provide a synopsis of the review teams’
`thoughts, deliberations and decisions associated with the CMC review of NDA 22-350,
`. Onglyza (saxagliptin) IR tablets. The intent is to capture the teams’ thoughts to facilitate
`future CMC supplement review and ONDQA discussions about regulatory submissions
`containing Quality by Design (QbD) information. This application was submitted as part
`of ONDQAs’ CMC Pilot Program, published in the Federal Register (Vol. 70, No.134,
`July 14, 2005).
`
`Initial evaluation of this NDA and the decision as to its fileability was conducted by the
`PAL assigned to this project, Su Tran. This Initial Qiality Assessment (IQA) review is
`filed in DFS/DARRTSI.
`
`In this initial assessment, several critical CMC deficiencies were identified and
`communicated to the applicant in the filing memo. The timely responses to these
`deficiencies provided critical CMC information to facilitate the full CMC evaluation of
`this NDA.
`
`The team review process for this specificpilot program application consisted of several
`independent reviews from each of the team members. These reviews were filedwithin
`the regulatory review timelines mandated under the Good Review Management
`Principles guidance. Each of these reviews focus on specific aspects of the CMC review,
`specific to the area of expertise of the individual reviewer. These reviews have been
`entered into DFS/DARRT82’3’4:
`
`Each of these reviews resulted in several CMC deficiencies which were communicated to
`the applicant. The evaluations of the responses to these deficiencies have also been
`entered into DFS/DARRTSS’6’7.
`
`The current document summarizes the pertinent issues discussed in the six individual
`reviews filed in DFS/DARRTS.
`
`II. Description of the Drug Product(s) and Drug Substance(s)
`
`' Initial Quality Assessment: Su Tran (chem.IQA/filing,27-AUG-2008)
`2 Drug substance CMC/QbD review: Prafull Shiromani (CMC Review 1, 28-OCT-2008)
`3 Drug product CMC/QbD review: Sharmista Chatterjee (Drug Product QbD Review, 24-NOV-2008)
`4 Overall CMC review: John Hill (CMC Review #1 for NDA 22-350, 08-DEC-2008)
`5 Drug substance CMC/QbD review: Prafull Shiromani (CMC Review 2, 23-MAR-2009)
`6 Drug product CMC/QbD review: Sharmista Chatterjee (Drug Product QbD Review, 23-MAR-2009)
`7 Overall CMC review: John Hill (Review, 23-MAR—2009)
`
`

`

`Drug Product
`
`‘ /'-—"‘ will be
`Saxagliptin film-coated tablets, 2.5 mg and 5 mg i’
`manufactured at the BMS facility in Mount Vernon, Indiana. Saxagliptin film
`coated tablets, 2.5 mg and 5 mg strength- \——— , have been
`developed for commercializationThe active in the drug product is the
`hydrochloride salt form of the drug substance. The structure for this salt form is
`provided as follows.
`
`11(4)
`
`H N+
`
`N
`
`- CI-
`
`CN
`
`Molecular Formula: C18H25N3Oz * HCl
`
`Molecular Weight: 351.87
`
`Since subjecting saxagliptin to common pharmaceutical operations such as
`
`M4)
`
`31(4)
`
`11(4)
`
`

`

`”(4)
`
`M4)
`
`M4)
`
`Based on tablet dimension measurements, saxagliptin film coated tablets, 2.5 mg,
`and saxagliptin film coated tablets, 5 mg, are expected to have an average
`thickness of 4.2 mm and average diameter of 8.2 mm.
`
`The saxagliptin film coated tablets contain the following excipients: lactose
`monohydrate, microcrystalline cellulose, croscarmellose sodium, magnesium
`state,
`.——————-
`yellow (2.5 mg),/’ pink (5 mg tablet),
`.
`_____. No novel excipients are used in the
`manufacture of saxagliptin 1 m coated tablets. Due to the M
`system, traditional excipient compatibility studies were not performed for the
`saxagliptin drug substance, it was addressed through saxagliptin stability studies.
`
`The two strengths will be differentiated by color and printing on the tablets.
`
`-
`
`o
`
`2.5 mg tablet, containing 2.79 mg saxagliptin hydrochloride (anhydrous):
`Round, bi-convex yellow tablet printed with “2.5” and “4214”
`5 mg tablet, containing 5.58 mg saxagliptin hydrochloride (anhydrous):
`Round, bi-convex pink tablet printed with “5’ and “4215”
`
`Saxagliptin film coated tablets will be packaged and marketed as 30 and 90 count
`in 95 mL and 500 count in 200 mL white, opaque,
`---——-"""—'—"“
`”T 7": bottles, with a two piece child resistant closure having an aluminum foil
`induction seal. Each bottle contains a cotton coil and one 2g pouch containing
`silica gel (desiccant) and activated carbon. 5 mg saxagliptin film coated tablets
`will also be packaged and marketed in aluminum/aluminum (alu/Alu) blisters.
`
`The quality of the drug product is assessed by visual and ’~ High
`Performance Liquid Chromatography (’-HPLC) methods and assured by
`Quality by Design (QbD) principles.
`
`Saxagliptin is a highly potent, selective, reversible, and competitive dipeptidyl
`.peptidase—4 (DPP4) inhibitor. DPP4 is the enzyme primarily responsible for the
`inactivation of the incretin hormones glucagon-like peptide-l (GLP-1) and
`glucose-dependent insulinotropic polypeptide (GIP). Incretin hormones are
`gastrointestinal hormones that increase insulin secretion in response to enteral
`stimulation. These hormones contribute to the control of postprandial glucose
`excursions in a glucose dependent manner, mitigating the risk of hypoglycemia.
`In addition to enhancing postprandial insulin release, GLP-l also reduces
`glucagon release from the pancreatic (Jr-cells, thereby reducing hepatic glucose
`production. This effect is also glucose-dependent, such that when plasma glucose
`is normal or low, the counter-regulatory response of glucagon release is not
`impaired.
`
`Saxagliptin is intended to improve glycemic control for patients with T2DM:
`- as monotherapy as an adjunct to diet and exercise;
`
`

`

`- in combination with metformin, a thiazolidinedione (TZD), or a
`sulfonylurea (SU)when the single agent alone, with diet and exercise does
`not provide adequate glycemic control; and
`‘
`- as initial combination with metformin, as an adjunct to diet and exercise,
`when treatment with dual saxagliptin and metformin therapy is
`appropriate.
`
`The proposed usual clinical dose is 5 mg once daily. The recommended dose is
`2.5 mg once daily in subjects with moderate or severe renal impairment, and end-
`stage renal disease requiring hemodialysis.
`
`Drug substance
`
`Saxagliptin drug substance will be manufactured at the BMS facility in Swords,
`Ireland using a /-— step commercial process fronV‘ starting materials. The
`A” , is used in the manufacture of
`saxagliptin tablets.
`
`M4)
`
`The Drug substance is saxagliptin. The formal chemical name is (lS,3S,5.S’)—2-
`[(23)-2-Amino—2-(3-hydr0xytricyclo [3 .3 . l . 1 3,7]dec-1 —y1)acetyl]-2-
`azabicyclo[3.l .0]hexane-3-carbonitrile, monohydrate. The molecular structure is
`provided as follows:
`
`no
`
`HzN
`
`I H20
`
`N
`
`CN
`
`The molecular formula is: C13H25N302 - H20 with a calculated molecular weight
`of 333.43.
`
`Sanapliptin has been chemically and structurally characterized using: P‘ ‘
`
`
`
`Saxagliptin drug substance is manufactured using a' / step commercial process
`from /starting materials. The lm is used in the
`manufacture of saxagliptin tablets.
`
`I1(4)
`
`

`

`Based on the 32-month primary stability data for saxagliptin drug substance from
`Process C and the lZ-month primary stability data for saxagliptin drug substance
`from Process D stored at 5°C (13°C), a ’ [period for the drug
`substance is granted with the following label statements:
`
`\
`
`III. Background
`
`The Quality section of the NDA was submitted as part of the FDA’s CMC Pilot Program.
`On June 9, 2006 EMS requested acceptance into the Agency’s CMC pilot program. BMS
`met with the Agency on August 14, 2006, to outline the saxagliptin QbD program and
`was accepted into the program on September 5, 2006. In the meeting minutes, FDA
`indicated BMS should submit a comprehensive Quality Overall Summary (QOS) and an
`expanded Pharmaceutical Development Section. BMS met with the Agency on April 26,
`2007 to further discuss BMS’s progress in the saxagliptin QbD development program.
`FDA agreed to BMS’s proposed NDA content, which would include a comprehensive
`QOS and an expanded Section 3.2.P.2 “Pharmaceutical Development.”
`
`The FDA provided the following in response dated November 18, 2005 to a CMC-
`specific End of Phase II Briefing Document, submitted August 22, 2005 (IND
`63,634,Serial No. 0085):
`
`would be acceptable
`o The starting materials
`provided appropriate technical information as outlined in the Agency’s reply is
`provided.
`0 Acceptance of the drug substance long term stability protocol.
`0 The Agency suggested the utilization of protocols to address changes to vendors,
`manufacturing processes, impurity profiles and acceptance specifications for
`starting materials, as well as changes to the manufacturing process and acceptance
`specifications for bulk drug substance.
`0 Acceptance of the proposed dissolution methodology which utilizes USP
`Apparatus II at 50 RPM in 0.1 N HCl.
`0 The Agency recommended modifying the drug product long term stability
`protocol for bottles, and accepted the proposed bracketing for blisters. The agency
`recommended modifying the proposed matrix to include testing each lot at the
`final time point.
`
`BMS submitted additional information on April 12, 2007 (IND 63,634 Serial No. 0189)
`on the proposed starting materials, along with information about the proposed
`commercial drug substance manufacturing process and our proposal to utilize data from
`both Process C and Process D to determine the drug substance re-test period. Also, BMS
`requested comments on a proposed change to the dissolution method approved during
`
`

`

`review of the End of Phase II briefing document. The Agency’s response received via
`electronic mail on May 3, 2007 and May 11, 2007 indicated the following:
`
`0 The proposal to base the retest period on long-term stability data obtained on both
`Process C and D was acceptable
`0 The proposed modification to the dissolution method was acceptable
`0 The Agency had no objection to the information provided on starting materials.
`0 The Agency requested BMS address the possibility of low levels of the
`"_'—_’
`
`in the drug substance.
`0 Discuss in the NDA observations regarding the behavior of saxagliptin tablets
`during dissolution testing.
`
`M4)
`
`12(4)
`
`BMS submitted Pre-NDA CMC questions to the Agency on December 27, 2007 (IND
`63,634 Serial No. 0238). A response to these questions was received via electronic mail
`on February 15, 2008. The FDA had no comments on BMS’ plan to submit 18- and 24-
`month LTSS data during the NDA review provided it was submitted no later than 6
`months after submission of the NDA. In addition, the Agency indicated that it would not
`be able to review a Post-Approval Management Plan as outlined by EMS, but that BMS
`had the option of submitting one or more comparability protocols permissible under the
`current draft guidance. As a result of these responses, BMS submitted a Comparability
`Protocol as part of the saxagliptin NDA.
`
`BMS submitted the NDA on June 30, 2008. Some of the issues identified in the Initial
`Quality Assessment1 were:
`
`0 Lack of characterization information on saxagliptin hydrochloride, the active
`ingredient form in the drug product
`
`0 Different dosage strengths having! .———-——-—————-——-——.W
`
`0 Lack of information ( 7AMr‘~——-“"—'——.
`
`0
`
`' 6"” not being proposed as a Critical Quality Attribute in the drug product
`manufacture ‘ ”m
`0 Use of2M
`
`Understanding and Design Space” studies
`0 Lack of Dissolution in the drug product specification
`
`h(4)
`
`l1(4)
`
`These and other issues identified in the IQA were addressed in subsequent reviewsz’z’4
`and adequately resolved.
`
`IV. Assessment of Implementation of Quality by Design
`
`As part of the pilot program, BMS was expected to include in this NDA an expanded
`Pharmaceutical Development section and critical CMC information that appropriately
`demonstrate product knowledge and process understanding of the drug substance and
`drug product using QbD principles and science-based approaches. In addition, a more
`
`

`

`comprehensive Quality Overall Summary (cQOS) summarizing all critical CMC
`elements, along with an evaluation and assessment of those elements was expected.
`
`These sections have been included in NDA 22-350. As part of the pilot program, the
`expanded pharmaceutical sections for both the drug substance and drug product have
`been independently assessed as part of ONDQA’s team review philosophy. Several
`deficiency letters have been communicated to EMS and their responses have been
`received and evaluated.
`
`The six reviews filed in DFS for this application 2’3’4’5’6’7 reflect the teams’ evaluation of
`the overall quality of the CMC data provided. Each review reflects the strengths and
`expertise of the individual team members. While these evaluations have been filed
`independently, significant team discussions about the adequacy of the CMC data
`submitted have provided a unifying focus to the review process. The result is an in-depth
`evaluation of the overall QbD data (both drug substance and drug product) which BMS
`has provided in support of this application and the applicability of these data to aspects of
`the general chemistry and stability of saxagliptin.
`
`After thorough evaluation of the quality data presented in this NDA, it is clear that BMS
`has demonstrated an understanding and implementation of aspects of QbD. The review
`process has also. identified areas in which BMS would benefit from further exploration,
`development and refinement of their design space.
`
`In general, the positive aspects of this QbD application have been identified as:
`
`0 The depth of specific information describing the control of the starting materials
`‘ ‘M—fl md
`
`the manufacturing of the drug substance (DS). This information includes
`- acceptable rationale for proven acceptable ranges (PARS) of reaction parameters
`and determination of the criticality of these parametersLPagM'g- ”- "2").
`
`o The implementation of suitable in-process PAT controls, e.g. 'a
`W (above
`reference: page 10).
`
`o The implementation ofQbD during dexielopment ofthe DS manufacturing
`process, facilitating the identification 0 critical and key process parameters.
`Critical Quality Attributes have also been identified (above reference: pages 45, 46, 52).
`
`0 The establishment of proven acceptable ranges (PARS) and normal operating
`ranges (NORs) to ensure consistent quality and yield of the DS. It is noteworthy
`that the NORs for the manufacture of the drug substance are within the
`experimental range studied in the DOE for the drug substance. All the
`experiments performed in this DOE produced high—quality drug substance thus,
`substantiating these NORs, i.e. the Design Space (above reference: pages 54,57).
`
`

`

`Rationale for selection of formulation variables e.g. coating thickness, choice of
`coating material, ratio of drug substance to p, in the coating suspension, pH
`of coating suspension3.
`
`M4)
`
`Presentation of scientific rationale for designating various drug product (DP)
`quality attributes as critical3.
`
`Implementation of a scientific, top down risk assessment methodology as per ICH
`Q9, e.g. use of '/"" diagrams, to determine operations that were critical to
`drug product quality. Once the critical operation was identified, risk assessment
`methodology was again used to determine the critical sub steps. The critical sub
`steps were then examined in detail using a combination of risk assessment and
`DOE based experimentation3.
`
`M4)
`
`Following a step by step rationale approach to determine DP manufacturing
`design space for critical variables at pilot scale based on DOE data and then scale
`to commercial scale3.
`‘
`
`approach to leverage understanding gained from
`Use of w————/‘
`experimentation related to DP manufacturing and use it to limit future trial and
`error type experiments.
`
`Presenting a rationale approach to use the
`a new operating range within/outside the design space3.
`
`
`
`model to select
`
`Points where BMS would benefit from further exploration and development include:
`
`Provide alclear thesis/rationale for why they decided to submit the additional
`QbD, DoE and expanded manufacturing data in the application and what their
`expectations were (what regulatory actions or relief were they seeking)
`
`The mathematical model and associated statistical analyses for the DOE
`associated with the manufacture of sazxagliptin were not presented/explained
`adequatelym the original submissionz. Preferably, these data should be presented
`in a systematic manner, with a few appropriate concise tables, e. g i) Independent
`(X) and Dependent (Y) variables for the particular DOE; ii) Experimental range-
`statistical and corresponding physical units of Xs; iii) Yates Algorithm - the value
`of Xs for each experimental batch - with the whole set of experimental batches
`manufactured in a random order so as minimize experimental error; iv) Table of
`Y- values for each batch with mean and SD; v). Mathematical model/equation
`generated by the computer from the input raw Ys' data in the form: A
`" 7"
`
`Correlation coefficients (R2)— to determine the fit ofeach
`g
`Y to the computer generated model; vii) Optimum Formulation- through
`computer search techniques (feasibility and grid searches); viii) Validation
`
`l1(4)
`
`11(4)
`
`

`

`experiment and it's data to confirm predictive power of the predictor equation; xi)
`Determination of critical Ys, i.e. Model Reduction/Refinement, by Principal
`Component Analysis or comparison of each regression coefficient with the
`standard error; x) Contour and Response Surface Plots, (in all fairness, this
`deficiency is not restricted to this BMS NDA, but to a few recent NDAs with
`QbD elements; this should be resolved, eventually, as the QbD paradigm
`becomes the norm in drug product development).
`
`Provide data justifying criticality analysis of excipients, i.e. to show if any
`variation in excipient properties would impact CQA of finished product. For
`example no data was presented to show if lot to lot variation in amounts
`.—--—-
`
`would have an impact on quality of drug
`product. This information was provided as a response to an information request
`letter6, response to comment #4, pg 6.
`
`Lumping several variables to assess probability (P), severity (S) and risk (R)
`while performing risk assessment for DP manufacturing process variables.
`Individual effects are thus masked. Upon communicating this deficiency to the
`sponsor, they a eed to follow the suggested approach for future submissions6’
`response to comment
`, pg 13)
`
`Lack of assurance of the controls in place to ensure content uniformity of finished
`drug product, especially for movements outside the target operating conditions
`within the design space, at commercial scale6’ “m”, P31).
`
`Some gaps identified in not evaluating interaction of design spaces. For example,
`sponsor did not evaluate interaction of PAR of nozzle parameter with variation in
`equipment design. They agreed to add appropriate manufacturing controls in the
`batch record in terms of nozzle dimensionsé’ “’5me ‘° ”men‘s” and 8’ pg 13’ 18).
`
`Lack of data presented in the original submission to show how proven acceptable
`ranges for critical coating process variables were scaled from pilot to commercial
`scale, for example no scale up correlations were resented. This was provided
`later in response to an information request letter ”65pm” t° °°mme“t#l4’ Pg 25).
`
`Minimal information was provided in the original submission about the in-process
`assay that is performed after at least ’——_——_~.‘—"
`u
`. Note, this in process step is performed only at
`commercial scale,A
`Similar in process step was not deemed necessary at pilot scale. Adequate
`information was provided about this assay in response to IR letters6’ ”pm” t° °°mm°m
`#16, 17,18 pg 28, 32-34)
`
`M4)
`
`It is opined that if this was well understood and controlled at all scales, such an in
`process step would not have been required at commercial scale. Having said that,
`
`10
`
`

`

`the in process step would give additional assurance that the final product would
`meet the criterion for assay.
`
`Lack of assurance in the original submission about procedures for handling
`movements within design space at commercial scale. In addition, some critical
`manufacturing procedures and controls including the design space table were not
`included in the P3 section. These omissions were corrected subsequentlsy via
`.
`1
`amendments to the NDA, 1n response to IR letters6’ ”Spmsm cmmems #3’ 4 ’
`6’ H’ 5 pg
`13-16, 19, 26-28)
`
`Insufficient information presented in the original submission for the DOE’s that
`were used for formulation and manufacturing process development (e.g. statistical
`analysis of the DOE data). These omissions were corrected subsequently via
`I‘CSponS6 to IR letters6’ response to comments #6, 21 pg 8, 36 respectively).
`
`I1(4)
`
`2
`
`1
`
`f
`
`11(4)
`
`< 5
`
`V. Notes for future inspections and Supplement Reviews
`
`1.) The applicant executed a statistically designed set of experiments (DOE) to test and
`verify the ranges proposed for key and critical process parameters of the/
`/
`1 process. The primary
`
`/purpose ofthe DOE was to confirm that the critical impurity/
`
`l_ was
`
`adequately controlled. Indeed, all/
`
`lexperiments, covering the extreme values of
`
`ll
`
`W“
`
`

`

`the parameters studied, produced drug substance with ~—- below the
`,7 reporting limit of the HPLC method. None of the main effects were
`significant, as determined by appropriate statistical tests, and hence, it was assumed
`that the associated f were insignificant. The applicant found that
`the quality of the drug substance1n each of these experiments was indistinguishable
`from the quality of the drug substance produced at commercial manufacturing scale.
`Based on the DOE results and the alignment between the DOE and commercial
`manufacturing batch quality, BMS believes that the design space proposed for the
`drug substance manufacture has been substantiated, to which this reviewer concurs.
`Accordingly, movement within the design5space requires no agency notification
`during routine commercial manufacturings.
`
`2.) One interesting feature in the developmental report for the saxaglintin drug nrodnct
`
`\
`
`\
`
`/
`
`\
`
`K
`
`\/"
`
`.
`
`b”)
`
`h(4)
`
`3.) A lingering concern is the issue of demonstration of adequate tablet content
`uniformity when moving outside the target operating condition but within the design
`space for active coating of tablets ’—"—" 1‘0 be noted that this is a
`relatively low dose drug (<2.5%) and USP<905> is currently followed to demonstrate
`content uniformity. The concern is based in part on the fact that it is noted that there
`is an increase in /" when moving outside the target operating condition (refer
`table 13. 1'1n amendment # 17 to the NDA dated January 21, 2009 )3. It1s
`recommended to evaluate the sponsor’s quality system to ensure that it includes
`appropriate sampling to assure content uniformity when moving outside the target
`operating condition but within the design space at commercial scale.
`
`bkfi
`
`4.) If the opportunity arises, participation by an ONDQA reviewer in any inspection to
`support a Prior Approval Supplement dealing with modification of the approved
`
`12
`
`

`

`design space is recommended. Onsite inspection by the reviewer would be beneficial
`to the investigator to understand the various approaches to QbD.
`
`5.) In the NDA application, BMS did not include dissolution as a regulatory requirement
`for final drug product release or stability. In order to attain approval of their
`application, BMS has agreed to include a regulatory test for dissolution for release
`and stability. The Agency indicated that we were open to discussions about
`alternative tests for dissolution/bioavailability in the future.7. The upshot of this
`action is that there will be further interactions/meetings with the applicant to explore
`alternative method(s) to evaluate dissolution. Such discussions will require
`participation by ONDQAs’ Biopharrn review staff.
`
`6.) The stability data for the saxagliptin 2.5 mg coated tablet packaged1n blisters
`—'—‘
`demonstrate that this packagingis not as robust as the Alu/Alu blister or
`the /bottle configuration. The stability data demonstrate that thereis a humidity
`and temperature affect on overall impurity profile; elevations1n either lead to the
`drug product failing to pass the proposed stability specification4. In the 74 day filing
`letter (12-SEP-2008), BMS was asked to Clarify whether the 2.5 mg tablets will be
`packaged in blisters because this packaging is not in the proposed labeling even
`though this packaging is listed for this dosage strength in the Container Closure and
`Stability sections of the NDA. In their response dated 15-OCT—2008, EMS states that
`they plan to launch the 2.5 mg tablets1n bottles. Subsequently, blister packs might be
`added for hospital unit doses and physician samples.7 Potentially, there will be a
`CMC supplementin the future requesting review and _127-“
`
`.7
`In such a case, the impact of the proposed blister
`material (moisture permeability, etc.) will be an important review consideration
`
`”(4)
`
`M4)
`
`l3
`
`

`

`This is a representation of an electronic record that was signed electronically and
`this page is the manifestation of the electronic signature.
`
`John C. Hill
`4/3/2009 10:24:03 AM
`CHEMIST
`
`Suong Tran
`4/3/2009 10:32:

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