Top Banner

of 62

rnd process.pdf

Apr 14, 2018

Download

Documents

Mostofa Rubal
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
  • 7/29/2019 rnd process.pdf

    1/62

    Compliant Formulation Development The

    Key to Successful Pharma Development

    Obergeri, May 4th 2012

    Dr. R. Rogasch

  • 7/29/2019 rnd process.pdf

    2/62

    2

    Regulatory Requirements in Formulation Development

    EU Scientifc Guidance Documents EMA (Clinical, CMC, Procedural)

    EP7 General Chapters, Monographs

    US FDA Guidance Documents (Clinical, CMS, Procedural)

    USP General Chapters and Methods (Dissolution Method Development, IVIVC

    requirements, Statistical Methodology)

    ICH Q8/Q9/Q10

  • 7/29/2019 rnd process.pdf

    3/62

    3

    Regulatory Requirements in Formulation Development

    EU Scientifc Guidance Documents Formulation Development

    IMP Procedure (pre-clinical data, dossier submission requirements, clinical studies)

    CMC requirements (specifications, stability data, pre-process validation)

    Bioequivalence or Biowaiver approach

  • 7/29/2019 rnd process.pdf

    4/62

    4

    Regulatory Requirements in Formulation Development

    EU EP7 requirements Generic Drug Development - Legal status of monographs

    Monographs are official standards

    The Convention on the Elaboration of a European Pharmacopoeia makes the texts of thePh. Eur. mandatory in all signatory parties

    The pharmaceutical legislation in the European Union

    makes monographs obligatory standards

    (2001/83/EC, 2001/81/EC)

    Monographs may be accepted as suitable standardseven when not obligatory

  • 7/29/2019 rnd process.pdf

    5/62

    5

    Regulatory Requirements in Formulation Development

    EU EP7 requirements Generic Drug Development - example

    Do Ph. Eur. specifications apply throughout shelf-life?

    A: Yes, specifications apply until time of use for raw materials and throughout period of

    validity for preparations

    B: No, Ph. Eur. requirements are for release only

    From ICH Quality Implementation Working Group - Integrated Implementation Training Workshop

    Breakout D: Pharmacopoeial Requirements, Kuala Lumpur, July 2010

  • 7/29/2019 rnd process.pdf

    6/62

    6

    Regulatory Requirements in Formulation Development

    EU EP7 requirements Generic Drug Development - example

    Do Ph. Eur. specifications apply throughout shelf-life?

    A: Yes, specifications apply until time of use for

    raw materials and throughout period of validity for Preparations (EP7, general notices)

    B: No, Ph. Eur. requirements are for release only.

    Implications : EP7 mongraph specifications (impurities) are shelf life indicating !

    From ICH Quality Implementation Working Group - Integrated Implementation Training Workshop

    Breakout D: Pharmacopoeial Requirements, Kuala Lumpur, July 2010

  • 7/29/2019 rnd process.pdf

    7/62

    7

    ICHQ8/9/10 Paradigm in Formulation Development

    Disclaimer

    The information within this presentation is based on the

    ICH Q-IWG members expert ise and experience, and

    represents the views of the ICH Q-IWG members for thepurposes of a training workshop.

  • 7/29/2019 rnd process.pdf

    8/62

    8

    QRM as part of development

    To assess the critical attributes of Raw materials

    Solvents

    Active Pharmaceutical Ingredient (API)

    Starting materials

    Excipients

    Packaging materials

    To establish appropriate specifications, identify criticalprocess parameters and establish manufacturing controls

    ICH Q9

  • 7/29/2019 rnd process.pdf

    9/62

    9

    II.3: QRM as part of development

    To decrease variability of quality attributes: reduce product and material defects

    reduce manufacturing defects

    To assess the need foradditional studies(e.g., bioequivalence, stability)

    relating to scale up and technology transfer

    To make use of the design space concept(see ICH Q8)

    ICH Q9

  • 7/29/2019 rnd process.pdf

    10/62

    10

    Key Steps for a product under Quality by Design (QbD)

    Product/Process Development

    Pharmaceutical

    Development

    PQS & GMP

    Local Environment

    Commercial Manufacturing

    Quality Unit (QP,..) level support by PQS

    Manage product lifecycle, inclu ding

    continual improvement

    Design Space (DS), RTR test ing

    Link raw material attributes and process parameters

    to CQAs and perform Risk Assessment Methodology

    Potential CQA (Critical Quality Attribute) identified &

    CPP (Critical Process Parameters) determined

    QTPP : Definition of intended use & produc tQuali ty Target

    Product Profile

    CPP : Crit ical

    Process Parameter

    CQA : Crit ical

    Quality Attribute

    Risk Management

    Opportunities

    Design to meet CQA using Risk Management &

    experimental stud ies (e.g. DOE)DOE : Design of Experiment

    Control Strategy

    Technology Transfer

    Batch Release

    Strategy

    Prior Knowledge (science, GMP,regulations, ..)

    Continual

    improvement

    Product/Process Understanding

    QRM principle apply at any stage

    Marketing Author isation

    Quality System PQS

  • 7/29/2019 rnd process.pdf

    11/62

    11

    P2 of CTD as part of a regulatory submission

    In line with Quality Risk Management ?

    EXAMPLE

  • 7/29/2019 rnd process.pdf

    12/62

  • 7/29/2019 rnd process.pdf

    13/62

    13

    Target Product Profile

    Drug substance properties; prior knowledge

    Proposed formulation and manufacturing process

    Determination of

    Cause Effect relationships(Risk Identification with subsequent Risk Analysis)

    Risk-based classification(Risk Evaluation)

    Parameters to investigate (e.g. by DOE)(Risk Reduction 1. proposal; 2. verified)

    FORMULATIONFORMULATION

    DESIGN SPACEDESIGN SPACEPROCESSPROCESS

    DESIGN SPACEDESIGN SPACE

    BY UNIT OPERATIONBY UNIT OPERATIONCONTROLCONTROL

    STRATEGYSTRATEGY

    Formulationunderst

    anding

    Formu

    lationunderst

    anding

    Pro

    cessunderstanding

    Pro

    cessundersta

    nding

    ReRe--evalu

    ationandconf

    irmation

    evalu

    ationandconf

    irmation

    ReRe--eval

    uationandcon

    firmation

    evaluationandcon

    firmation

    Product and process

    characteristics on the

    final drug product

    Review events

    DevelopmentDevel

    opm.

    Operat

    ion

    Research

    Phase 1

    Phase 2

    Phase 3

    Launch

  • 7/29/2019 rnd process.pdf

    14/62

    14

    Risk Review

    Risk Assessment

    Risk Evaluation

    unacceptable

    Risk Control

    Risk Analysis

    Risk Reduction

    Risk Identifi cation

    Review Events

    Risk Acceptance

    Initiate

    Quality Risk Management Process

    Output / Result of the

    Quality Risk Management Process

    RiskManagementto

    olsR

    iskCommunication

    Teamfocused

    Int

    ernalconsultation

    Stakeholderinvolvement

    Responsibilities in regulatory operations

    Industry

    A) Reviewers

    EXAMPLE

    B) Inspectorates

  • 7/29/2019 rnd process.pdf

    15/62

    15

    Formulation Strategies for Phase I/II Clinical Programs

    General Outline

    The overall sequence for DP development for each phase/clinical trial can be summarized as

    follows:

    Define the best formulation, with the choice of excipients based on maximizing the physical

    and chemical stability of the API

    Ensure the formulation provides the desired in vitro release of drug

    Conduct pharmacokinetic studies in animals, if models are available that are known to

    predict clinical responses.

    Define the best manufacturing process for DP

    Place the final DP prototype on accelerated stability in intended packaging

    Conduct GMP manufacture and packaging of clinical DP

    Generate batch release data and certificate of analysis (CoA) for clinical DP

    Initiate an accelerated stability program for clinical DP (batch made at full scale)

    Submit supporting formulation and analytical data as part of the regulatory filing to request

    approval (i.e., from the FDA, EU, etc.) for using the DP in a clinical study

  • 7/29/2019 rnd process.pdf

    16/62

    16

    Formulation Strategies for Phase I/II Clinical Programs

    General Considerations Oral Dosage Forms

    Material Property Assessment

    API (solubility, Polymorphism XRD etc.)

    PSD (DLS, LLD)

    Morphology (SEM)

    Compound Dissolution

    Flow/cohesion

    Powder compaction

    Hardness, tensile strength, brittel fracture index

    Excipient/API interactions

    Degradation Pathways

  • 7/29/2019 rnd process.pdf

    17/62

    17

    Formulation Strategies for Phase I/II Clinical Programs

    General Considerations Oral Dosage Forms

    Bioavailability Enhancement

    API (solubility enhancement)

    PSD (micronization)

    Solubility Screening, w/o partition

    Precipitation inhibition (API/surfactant/polymer combinations)

    Amorphous Dispersions (solid solutions, dispersion in polymer matrix)

    Coatings (multi-particulates in capsules)

    Lipid Systems (fat-matrix, SEDDS, SMEDDS, liposomal carrier)

  • 7/29/2019 rnd process.pdf

    18/62

    18

    Formulation Strategies for Phase I/II Clinical Programs

    IR-Oral Dosage Forms

    Capsule, Tablet (IR dosage forms)

    Direct compression

    Dry Granulation

    Wet Granulation

    Tabletting/Capsule Filling

    Film Coating

    Hot Melt Extrusion

  • 7/29/2019 rnd process.pdf

    19/62

    19

    Formulation Strategies for Phase I/II Clinical Programs

    CR - Oral Dosage Forms

    Capsule, Tablet (CR dosage forms)

    Matrix

    Multiparticulates

    Soft Gel Capsules

    Liquid filled Capsules

    Fuctional Film Coating

    Hot Melt Extrusion

    Osmotic Systems

  • 7/29/2019 rnd process.pdf

    20/62

    20

    Formulation Strategies for Phase I/II Clinical Programs

    Solid Orals - Excipients

    Unit dose to contain powders or

    controlled release pellets

    1%-5%Gelatin

    HPMC

    Polysaccharides

    Capsules

    Improves powder flow and prevents

    static charging

    Less than 1%Fumed silica

    Talc

    Glidants

    Tailors drug release rate10%-95%HPMC

    Polyethylene oxide

    Polyvinylpyrrolidone (PVP)

    Controlled

    release/matrix

    Aids in breakup of tablets or

    granules in aqueous media

    Less than 5%Sodium starch glycolate

    Croscarmellose sodium Crospovidone

    Disintegrants

    Prevents sticking of formulation to

    processing surfaces

    Less than 2%Magnesium stearate Stearic acid

    Glyceryl behenate

    Lubricants

    Provides strength in dry and wet

    processing of powders

    5%-10%Hydroxypropyl cellulose (HPC)

    HPMCPovidone

    Binders

    Imparts compressibility and tensile

    strength to tablets

    10%-95%Mannitol

    Microcrystalline cellulose Starch

    Ductile fillers

    Imparts hardness and strength to

    tablets

    10%-95%Lactose

    Calcium phosphate, dibasic

    Brittle fillers

    FunctionApproximate

    ranges (%)MaterialsExcipient type

    Formulation ExcipientsSolid Orals

  • 7/29/2019 rnd process.pdf

    21/62

    21

    Formulation Strategies for Phase I/II Clinical Programs

    Solid Orals - Excipients

    Cosmetic or controlled release coatings1%-30%HPMC

    Cellulose acetate , Ethylcellulose,

    Polymeric acrylates

    Coating ingredients Film

    polymers

    Cosmetic appearance, marketingLess than 2%Titanium dioxide, Iron oxides, Dyes and

    lakes

    Colorants

    Hide unpleasant drug taste, essential for

    chewable formulations

    1%-5%Sucrose

    Aspartame

    Mannitol

    Flavors

    Taste masking agents

    Mitigate chemical degradation,

    oxidation

    Less than 1%BHT/BHA

    Citric acid

    Chemical stabilizers

    Improve solubility and wettability of

    hydrophobic drugs and improve

    bioavailability

    0.5%-5%Poloxamer 407

    SLS

    CyclodextrinsHPMC and acid derivatives

    HPC

    Solubilizers, dispersants,

    precipitation inhibitors

  • 7/29/2019 rnd process.pdf

    22/62

    22

    Formulation Strategies for Phase I/II Clinical Programs

    Solid Orals - Excipients

    Improve processability, Prevent

    sticking

    Less than 1% Less

    than 0.5%

    Glycerol triacetate, Fatty acid salts,

    esters, Polyethylene glycol,

    Talc

    Plasticizers, Anti-tack

    agent

  • 7/29/2019 rnd process.pdf

    23/62

    23

    Formulation Strategies for Phase I/II Clinical Programs

    Parenteral Dosage Forms

    Parenteral/injectable Solutions (lyophilization)

    Colloidal Suspensions (peptides, proteins)

    Emulsions

    Liposomal Systems

    Suspensions

  • 7/29/2019 rnd process.pdf

    24/62

    24

    Formulation Strategies for Phase I/II Clinical Programs

    Liquid Orals/Parenterals Excipients

    Maintain osmolarity forparenterals, adjustviscosity, mechanicalstability for lyophilizedcakes

    Less than 10%Sodium chlorideHydroxypropylmethylcellulose(HPMC)Mannitol Dextrose

    Bulking agents

    Maintain pH for optimumsolubility, and comfortfor injectableformulations

    Enough foradjusting todesired pH

    Sodium chloride Sodiumacetate Sodiumphosphate (andcorrespondingacids) Sodium

    hydroxide

    Buffering agents

    Helps with poorly aqueoussoluble drugs20%-50%EthanolPolyethylene glycolPropylene glycolN-Methylpyrrolidone

    Cosolvents

    Main solubilizing/suspendingvehicle for allcomponents

    50%-90%WaterVegetable oils

    Polyethylene glycolPropylene glycol

    Diluent

    Functionpproximateranges (%)

    Materialsxcipient type

  • 7/29/2019 rnd process.pdf

    25/62

    25

    Formulation Strategies for Phase I/II Clinical Programs

    Oral Liquid/Parenteral - Excipients

    Sweeteners Masking of

    drug tast

    Less than

    2%

    Sucrose, aspartame

    Peppermint oil,

    flavors

    Flavoring/tastemasking

    Antioxidants, free radicalscavengers

    Less than2%

    Butylatedhydroxytoluene/anisole

    (BHT/BHA)

    Citric acid/citrate

    Chemical stabilizers

    Prevent microbial growthLess than

    2%

    Benzyl alcohol Methyl/

    propyl parabens

    Benzalkonium

    chloride Thimerosal

    Preservatives

    Bind metal impurities to

    prevent

    complexation and

    reactions

    Less than

    1%

    Edetate sodium (EDTA)

    Citric acid/citrate

    Chelating agents

    Improve drug solubility,

    emulsification,

    suspension of drugparticles, prevent

    precipitation

    Less than

    5%

    Hydroxypropyl-beta-

    cyclodextrin

    Sulfobutylether-beta-cyclodextrin

    HPMC

    Polaxamer 407

    Sodium lauryl sulfate

    (SLS)

    Phospholipids

    Cremophors

    Labrasol

    Vitamin E TPGS

    Solubilizers/surfactants

  • 7/29/2019 rnd process.pdf

    26/62

    26

    Project Case Study

    The information within this presentation is based

    on the ICH Q-IWG members expertise and experience,

    and represents the views of the ICH Q-IWG members

    for the purposes of a training workshop.

    Disclaimer

  • 7/29/2019 rnd process.pdf

    27/62

    27

    Outline of Presentation

    Key Steps for Quality by Design

    Case Study Organization

    Introducing API and Drug Product

    Discussion of concepts of Quality Target Product Profile, processes, composition

    Description of API & Drug Product process development Discussion of illustrative examples of detailed approaches from the case study

    Batch release

  • 7/29/2019 rnd process.pdf

    28/62

    28

    Purpose of Case Study

    Illustrative example

    Covers the concepts and integrated implementation of ICH Q8, 9 and

    10

    Not the complete content for a regulatory filing

    Note: this example is not intended to represent the preferred or

    required approach.

  • 7/29/2019 rnd process.pdf

    29/62

  • 7/29/2019 rnd process.pdf

    30/62

    30

    Basis for Development Information

    Fictional active pharmaceutical ingredient (API)

    Drug product information is based on the Sakura Tablet case study

    Full Sakura case study can be found at

    http://www.nihs.go.jp/drug/DrugDiv-E.html

    Alignment between API and drug product API Particle size and drug product dissolution

    Hydrolytic degradation and dry granulation /direct compression

    http://www.nihs.go.jp/drug/DrugDiv-E.htmlhttp://www.nihs.go.jp/drug/DrugDiv-E.html
  • 7/29/2019 rnd process.pdf

    31/62

  • 7/29/2019 rnd process.pdf

    32/62

    32

    Quality attr ibute focus

    Technical Examples

    API

    Drug Product

    CompressionReal Time

    Release testing(Assay, CU, Dissolution)

    BlendingAPI

    Crystallization

    - Final crystallization step

    - Blending

    - Direct compression

    - Particle size control

    - Assay and content uniformity

    - Dissolution

    Process focus

  • 7/29/2019 rnd process.pdf

    33/62

    33

    Process Step Analysis

    For each example

    Risk assessment

    Design of experiments

    Experimental planning, execution & data analysis

    Design space definition

    Control strategy

    Batch release

    Design of

    ExperimentsDesign

    Space

    Control

    StrategyBatch

    ReleaseQRM

  • 7/29/2019 rnd process.pdf

    34/62

    34

    QbD Story per Unit Operation

    Process

    Variables

    Design of

    Experiments

    Quality

    Risk Management

    Illustrative Examples of Unit Operations:

    QTPP

    & CQAs

    Design

    Space

    Control

    StrategyBatch

    Release

    CompressionReal Time

    Release testing(Assay, CU, Dissolution)

    BlendingAPI

    Crystallization

  • 7/29/2019 rnd process.pdf

    35/62

    35

    Introducing API and Drug Product

  • 7/29/2019 rnd process.pdf

    36/62

    36

    Assumptions & Prior Knowledge

    API is designated as Amokinol

    Single, neutral polymorph

    Biopharmaceutical Classification System (BCS) class II low solubility & high permeability

    API solubility (dissolution) affected by particle size Crystallization step impacts particle size

    Degrades by hydrolytic mechanism Higher water levels and elevated temperatures will increase degradation

    Degradates are water soluble, so last processing removal point is the aqueous extraction step

    Degradates are not rejected in the crystallization step

    In vitro-in vivo correlation (IVIVC) established allows dissolution to be used as

    surrogate for clinical performance

    Drug product is oral immediate release tablet

  • 7/29/2019 rnd process.pdf

    37/62

    37

    Quality Target Product Profile (QTPP)Safety and Efficacy Requirements

    Appearance, elegance, size,

    unit integrity and other characteristics

    No off-taste, uniform color,

    and suitable for global marketSubjective Properties

    Hydrolysis degradation & dissolution changescontroll ed by packaging

    Degradates below ICH or to be qualified and nochanges in bioperformance over expiry period

    Chemical and Drug Product Stability: 2year shelf life (worldwide = 30C)

    Acceptable API PSD

    Dissolution

    PSD that does not impact bioperformance or pharm

    processing

    Patient efficacy Particle

    Size Distribution (PSD)

    Accept able hydrolysis degradate levels at release,

    appropriate manufacturing environment controls

    Impurities and/or degradates

    below ICH or to be qualifiedPatient Safety chemical purity

    Identity, Assay and Uniformity30 mgDose

    Translation into

    Quality Target Product Profile (QTPP)Characteristics / RequirementsTablet

    QTPP may evolve during li fecycle during development and commercial manufacture - as new knowledge is

    gained e.g. new patient needs are identified, new technical information is obt ained about the product etc.

  • 7/29/2019 rnd process.pdf

    38/62

  • 7/29/2019 rnd process.pdf

    39/62

    39

    Tablet Formulation

    Pharmacopoeial

    or othercompendial

    specification

  • 7/29/2019 rnd process.pdf

    40/62

    40

    Drug Product Process

    Blending

    Lubrication

    Compression

    Film coating

    API and ExcipientsAmokinol

    D-mannitol

    Calcium hydrogen phosphate hydrate

    Sodium starch glycolateLubricantMagnesium Stearate

    CoatingHPMCMacrogol 6000

    titanium oxide

    iron sesquioxide

  • 7/29/2019 rnd process.pdf

    41/62

  • 7/29/2019 rnd process.pdf

    42/62

    42

    Overall Risk Assessment for Process

    Cou

    pling

    Rea

    ction

    Aqu

    eous

    Ex

    tra

    ctions

    Distillative

    So

    lven

    tSw

    itc

    h

    e

    m

    -

    Cont

    inuous

    Crsta

    lliza

    tion

    Cen

    trifuga

    l

    Filtra

    tion

    Ro

    tary

    Dry

    ing

    Manu

    fac

    ture

    Mo

    istur

    eCon

    tro

    l

    Blen

    ding

    Lu

    brica

    tion

    Comp

    ress

    ion

    Coa

    ting

    Pack

    ag

    ing

    in vivo performance*

    Dissolution

    Assay

    Degradation

    Content Uniformity

    Appearance

    Friability

    Stability-chemical

    Stability-physical

    Drug Substance Drug Product

    * includes bioperformace of API, and

    safety(API purity)

    additional study required

    known or potential impact to CQA

    known or potential impact to CQA

    current controls mitigate risk

    no impact to CQA

    Process Steps

    CQA

    Example from Case Study

  • 7/29/2019 rnd process.pdf

    43/62

    43

    Overall Risk Assessment for Process

    Cou

    pling

    Rea

    ction

    Aqu

    eous

    Ex

    tra

    ctions

    Distillative

    So

    lven

    tSw

    itc

    h

    e

    m

    -

    Cont

    inuous

    Crsta

    lliza

    tion

    Cen

    trifuga

    l

    Filtra

    tion

    Ro

    tary

    Dry

    ing

    Manu

    fac

    ture

    Mo

    istur

    eCon

    tro

    l

    Blen

    ding

    Lu

    brica

    tion

    Comp

    ress

    ion

    Coa

    ting

    Pack

    ag

    ing

    in vivo performance*

    Dissolution

    Assay

    Degradation

    Content Uniformity

    Appearance

    Friability

    Stability-chemical

    Stability-physical

    Drug Substance Drug Product

    * includes bioperformace of API, and

    safety(API purity)

    additional study required

    known or potential impact to CQA

    known or potential impact to CQA

    current controls mitigate risk

    no impact to CQA

    Process Steps

    CQA

  • 7/29/2019 rnd process.pdf

    44/62

    44

    API Semi-Continuous Crystallization

    Designed to minimize hydrolytic degradation (degradate belowqualified levels)

    Univariate experimentation example

    FMEA of crystallization process parameters

    High risk for temperature, feed time, water level

    Test upper end of parameter ranges (represents worst case) with variation inwater content only and monitor degradation

    Proven acceptable upper limits defined for above parameters

    Note that in this case study, the distillative solvent switch prior to crystallizationand crystallization itself are conducted at lower temperatures and no degradationoccurs in these steps

  • 7/29/2019 rnd process.pdf

    45/62

    45

    API Semi-Continuous Crystallization

    Designed to control particle size

    Multivariate DOE example leading to predictive model

    FMEA of parameters using prior knowledge

    High risk for addition time, % seed, temperature, agitation

    DOE: half fraction factorial using experimental ranges based on QTPP,operational flexibility & prior knowledge

    Design space based on predictive model obtained by statistical analysis ofDOE data

    Particle size distribution (PSD) qualified in formulation DOE anddissolution studies

  • 7/29/2019 rnd process.pdf

    46/62

    46

    Risk Assessment:Particle Size Distribution (PSD) Control

    What is the Impact that ------------- will have on PSD? 1) minimal5) moderate9) significantWhat is the Probability that variations in ------------ will occur? 1) unlikely5) moderately likely 9) highly likely

    What is ourAbil ity to Detect a meaningful variation in --------------- at a meaningful control point? 1) certain5) moderate 9) unlikely

    Unit Operation Parameter IM

    PACT

    PROB

    .

    Dete

    ct

    RPN

    Comments

    Crystallization Feed Temperature 1 5 1 5

    Prior knowledge (slowness of crystallization kinetics) ensures that thehot crystallizer feed will be well dispersed and thermally equilibrated

    before crystallizing. Hence no impact of feed temp variation oncrystal size.

    Crystallization Water content of Feed 1 5 5 25Prior knowledge (solubility data) shows that small variations in waterdo not affect crystalliation kinetics.

    Crystallization Addition Time (Feed Rate) 9 5 9 405

    Fast addition could result in uncontrolled crystallization. Detection of

    short addition time could occur too late to prevent this uncontrolledcrystallization, and thus impact final PSD.

    Crystallization Seed wt percentage 9 5 5 225Prior knowledge (Chemical Engineering theory) highlights seed wtpercentage variations as a potential source of final PSD variation

    Crystallization Antisolvent percentage 1 1 1 1

    Yield loss to crystallization already low (< 5%), so reasonable

    variations in antisolvent percentage (+/- 10%) will not affect the

    percent of batch crystallized, and will not affect PSD

    Crystallization Temperature 9 5 9 405Change in crystallization temperature is easily detected, but ratedhigh since no possible corrective action (such as, if seed has been

    dissolved)

    Crystallization Agitation (tip speed) 9 5 5 225Prior knowledge indicates that final PSD highly sensitive to Agitation,

    thus requiring further study.

    Crystallization Seed particle size distribution 9 1 1 9Seed PSD controlled by release assay performed after air attrition

    milling.

    Crystallization Feed Concentration 1 1 1 1 Same logic as for antisolvent percentage

    To be investigated

    in DOE

  • 7/29/2019 rnd process.pdf

    47/62

    47

    Options for Depicting a Design Space

    Large square represents the ranges tested in the DOE.

    Red area represents points of failure

    Green area represents points of success.

    Oval = full design space represented

    by equation

    Rectangle represent ranges Simple, but a portion of the design

    space is not utilized

    Could use other rectangles within oval

    Exact choice of above options can bedriven by business factors

    Temperature

    Pressure

    For purposes of this case study, an acceptable design space based on ranges was chosen

    Seedwt%

    API C t lli ti

  • 7/29/2019 rnd process.pdf

    48/62

    48

    API Crystallization:

    Design Space & Control Strategy

    Control Strategy should address:

    Parameter controls

    Distillative solvent switch achieves target water content

    Crystallization parameters are within the design space

    Testing

    API feed solution tested for water content

    Final API will be tested for hydrolysis degradate

    Using the predictive model, PSD does not need to be routinely tested since it is

    consistently controlled by the process parameters

  • 7/29/2019 rnd process.pdf

    49/62

  • 7/29/2019 rnd process.pdf

    50/62

    50

    Drug Product

    Immediate release tablet containing 30 mg Amokinol

    Rationale for formulation composition and process selection provided

    In vitro-in vivo correlation (IVIVC) determination

    Correlation shown between pharmacokinetic data and dissolution results

    Robust dissolution measurement needed

    For a low solubility drug, close monitoring is important

    D P d t Di t C i M f t i

  • 7/29/2019 rnd process.pdf

    51/62

    51

    Drug Product Direct Compression Manufacturing

    Process

    Focus of

    Story

    Example from Case Study

    Lubrication

  • 7/29/2019 rnd process.pdf

    52/62

    52

    Initial Quality Risk Assessment

    Impact of Formulation and Process unit operations on Tablet CQAsassessed using prior knowledge

    Also consider the impact of excipient characteristics on the CQAs

    Drug

    substance

    particle size

    Moisture

    content in

    manufacture

    Blending Lubrication Compression Coating Packaging

    - Low risk

    - Medium risk

    - High risk

    Degradation

    Content uniformity

    Appearance

    Friability

    Stability-chemical

    Stability-physical

    in vivoperformance

    Dissolution

    Assay

    Example from Case Study

  • 7/29/2019 rnd process.pdf

    53/62

  • 7/29/2019 rnd process.pdf

    54/62

    54

    Predictive Model for DissolutionA mathematical representation of the design space

    Batch 1 Batch 2 Batch 3

    Model prediction 89.8 87.3 88.5

    Dissolution testing result92.8

    (88.494.2)

    90.3

    (89.0-102.5)

    91.5

    (90.5-93.5)

    Prediction algorithm:

    Diss = 108.9 11.96 API 7.55610-5 MgSt 0.1849 LubT

    3.78310-2 Hard 2.55710-5 MgSt LubT

    Factors include: API PSD, lubricant (magnesium stearate) specific

    surface area, lubrication time, tablet hardness (via compression force)Confirmation of model

    Example from Case Study

    Continue model verification with dissolution testing of production material, as needed

  • 7/29/2019 rnd process.pdf

    55/62

  • 7/29/2019 rnd process.pdf

    56/62

    56

    Drug Product CQA -

    Assay & Content Uniformity Summary

    Quality r isk assessment Potential impact for API particle size, moisture control, blending, and lubrication

    Moisture will be controlled in manufacturing environment

    Consider possible control strategy approaches Experimental plan to develop design space using input material and process factors

    In-process monitoring

    Assay assured by weight control of tablets made from uniform powder

    blend with acceptable API content by HPLC Blend homogeneity by on-line NIR to determine blending endpoint, includes feedback loop

    API assay in blend tested by HPLC

    Tablet weight by automatic weight control with feedback loop

  • 7/29/2019 rnd process.pdf

    57/62

    57

    Blending Process Control Options

    Decision on conventional vs. RTR testing

    Example from Case Study

    P C t l O ti 2

  • 7/29/2019 rnd process.pdf

    58/62

    58

    Process Control Option 2Blend uniformity monitored using a process analyser

    On-line NIR spectrometer used to

    confirm scale up of blending

    Blending operation complete when

    mean spectral std. dev. reaches

    plateau region Plateau may be detected using statistical

    test or rules

    Feedback control to turn off blenderCompany verifies blend does not

    segregate downstream Assays tablets to confirm uniformity

    Conducts studies to try to segregate API

    0

    0.005

    0.01

    0.015

    0.02

    0.025

    0.03

    0.035

    0.04

    0.045

    0 32 64 96 128

    Revolution (block number)

    meanspectralstan

    darddeviation

    Pilot Scale

    Full Scale

    Plateau region

    Number of Revolutions of Bl ender

    Data analysis model will be provided

    Plan for updating of model availableAcknowledgement: adapted from ISPE PQLI Team

    Example from Case Study

  • 7/29/2019 rnd process.pdf

    59/62

    59

    Batch Release Strategy

    Finished product not tested for assay, CU and dissolution

    Input materials meet specifications and are tested API particle size distribution

    Magnesium stearate specific surface area

    Assay calculation Verify (API assay of blend by HPLC) X (tablet weight)

    Tablet weight by automatic weight control (feedback loop), %RSD of 10 tablets

    Content Uniformity On-line NIR criteria met for end of blending (blend homogeneity)

    Tablet weight control results checked

    Dissolution Predictive model using input and process parameters calculates for each batch that dissolution meets

    acceptance criteria

    Input and process parameters used are within the filed design space Compression force is monitored for tablet hardness

    Water content NMT 3% in finished product (not covered in this case study)

  • 7/29/2019 rnd process.pdf

    60/62

    60

    Drug Product Specifications

    Use for stability, regulatory testing, site change, whenever RTR testing is not possible Input materials meet specifications and are tested

    API PSD

    Magnesium stearate specific surface area

    Assay calculation (drug product acceptance criteria 95-105% by HPLC) Verify (API assay of blend by HPLC) X (tablet weight)

    Tablet weight by automatic weight control (feedback loop) For 10 tablets per sampling point,

  • 7/29/2019 rnd process.pdf

    61/62

  • 7/29/2019 rnd process.pdf

    62/62

    Conclusions

    Better process knowledge is the outcome of QbD development

    Provides the opportunity for flexible change management

    Use Quality Risk Management proactively

    Multiple approaches for experimental design are possible

    Multiple ways of presenting Design Space are acceptable Predictive models need to be confirmed and maintained

    Real Time Release Testing (RTRT) is an option

    Opportunity for efficiency and flexibility