药品开发_第1页
药品开发_第2页
药品开发_第3页
药品开发_第4页
药品开发_第5页
已阅读5页,还剩38页未读 继续免费阅读

下载本文档

版权说明:本文档由用户提供并上传,收益归属内容提供方,若内容存在侵权,请进行举报或认领

文档简介

Product Development:Case Study Overview, ICH, November 2010,Disclaimer,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., ICH, November 2010,Outline of Presentation,Key Steps for Quality by DesignCase Study OrganizationIntroducing API and Drug Product Discussion of concepts of Quality Target Product Profile, processes, compositionDescription of API & Drug Product process development Discussion of illustrative examples of detailed approaches from the case studyBatch release, ICH, November 2010,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, including continual improvement,Design Space (DS), RTR testing,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 & product,Quality TargetProduct Profile,CPP : CriticalProcess Parameter,CQA : CriticalQuality Attribute,Risk Management,Opportunities,Design to meet CQA using Risk Management & experimental studies (e.g. DOE),DOE : Design of Experiment,Control Strategy,Technology Transfer,Batch ReleaseStrategy,Prior Knowledge (science, GMP, regulations, .),Continualimprovement,Product/Process Understanding,QRM principle apply at any stage,Marketing Authorisation,Quality System PQS, ICH, November 2010,Purpose of Case Study,Illustrative exampleCovers the concepts and integrated implementation of ICH Q8, 9 and 10Not the complete content for a regulatory filingNote: this example is not intended to represent the preferred or required approach., ICH, November 2010,Case Study Organization, ICH, November 2010,Basis for Development Information,Fictional active pharmaceutical ingredient (API) Drug product information is based on the Sakura Tablet case studyFull Sakura case study can be found at http:/www.nihs.go.jp/drug/DrugDiv-E.html Alignment between API and drug productAPI Particle size and drug product dissolutionHydrolytic degradation and dry granulation /direct compression, ICH, November 2010,Organization of Content,Quality Target Product Profile (QTPP)API properties and assumptionsProcess and Drug product composition overviewInitial risk assessment of unit operationsQuality by Design assessment of selected unit operations, ICH, November 2010,Quality attribute focus,Technical Examples,APIDrug Product,Compression,Real Time Release testing(Assay, CU, Dissolution),Blending,APICrystallization,- Final crystallization step,- Blending- Direct compression,- Particle size control,- Assay and content uniformity - Dissolution,Process focus, ICH, November 2010,Process Step Analysis,For each exampleRisk assessmentDesign of experimentsExperimental planning, execution & data analysisDesign space definitionControl strategyBatch release,Design ofExperiments,Design Space,Control Strategy,Batch Release,QRM, ICH, November 2010,QbD Story per Unit Operation,Process Variables,Design ofExperiments,QualityRisk Management,Illustrative Examples of Unit Operations:,QTPP & CQAs,Design Space,Control Strategy,Batch Release,Compression,Real Time Release testing(Assay, CU, Dissolution),Blending,APICrystallization, ICH, November 2010,Introducing API and Drug Product, ICH, November 2010,Assumptions,API is designated as AmokinolSingle, neutral polymorphBiopharmaceutical Classification System (BCS) class II low solubility & high permeabilityAPI solubility (dissolution) affected by particle sizeDegrades by hydrolytic mechanismIn vitro-in vivo correlation (IVIVC) established allows dissolution to be used as surrogate for clinical performanceDrug product is oral immediate release tablet, ICH, November 2010,Assumptions & Prior Knowledge,API is designated as AmokinolSingle, neutral polymorphBiopharmaceutical Classification System (BCS) class II low solubility & high permeabilityAPI solubility (dissolution) affected by particle sizeCrystallization step impacts particle sizeDegrades by hydrolytic mechanismHigher water levels and elevated temperatures will increase degradationDegradates are water soluble, so last processing removal point is the aqueous extraction stepDegradates are not rejected in the crystallization stepIn vitro-in vivo correlation (IVIVC) established allows dissolution to be used as surrogate for clinical performanceDrug product is oral immediate release tablet, ICH, November 2010,Quality Target Product Profile (QTPP)Safety and Efficacy Requirements,QTPP may evolve during lifecycle during development and commercial manufacture - as new knowledge is gained e.g. new patient needs are identified, new technical information is obtained about the product etc., ICH, November 2010,API Unit Operations,Coupling Reaction,Aqueous Extractions,Distillative Solvent Switch,Semi ContinuousCrystallization,Centrifugal Filtration,Rotary Drying,Coupling of API Starting Materials,Removes water, prepares API for crystallization step,Addition of API in solution and anti-solvent to a seed slurry,Filtration and washing of API,Drying off crystallization solvents,Removes unreacted materials. Done cold to minimize risk of degradation,Understand formation & removal of impurities,Example from Case Study, ICH, November 2010,Tablet Formulation, ICH, November 2010,Drug Product Process,Blending,Lubrication,Compression,Film coating,API and ExcipientsAmokinolD-mannitolCalcium hydrogen phosphate hydrateSodium starch glycolate,LubricantMagnesium Stearate,CoatingHPMC,Macrogol 6000titanium oxideiron sesquioxide, ICH, November 2010,Overview of API and Drug Product Case Study ElementsRepresentative Examples from the full Case Study, ICH, November 2010,Overall Risk Assessment for Process,Process Steps,CQA,Example from Case Study, ICH, November 2010,Overall Risk Assessment for Process,Process Steps,CQA, ICH, November 2010,API Semi-Continuous Crystallization,Designed to minimize hydrolytic degradation (degradate below qualified levels)Univariate experimentation exampleFMEA of crystallization process parametersHigh risk for temperature, feed time, water levelTest upper end of parameter ranges (represents worst case) with variation in water content only and monitor degradationProven acceptable upper limits defined for above parametersNote that in this case study, the distillative solvent switch prior to crystallization and crystallization itself are conducted at lower temperatures and no degradation occurs in these steps, ICH, November 2010,API Semi-Continuous Crystallization,Designed to control particle sizeMultivariate DOE example leading to predictive modelFMEA of parameters using prior knowledgeHigh risk for addition time, % seed, temperature, agitationDOE: half fraction factorial using experimental ranges based on QTPP, operational flexibility & prior knowledgeDesign space based on predictive model obtained by statistical analysis of DOE dataParticle size distribution (PSD) qualified in formulation DOE and dissolution studies, ICH, November 2010,Risk Assessment: Particle Size Distribution (PSD) Control,To be investigatedin DOE, ICH, November 2010,Options for Depicting a Design Space,Large square represents the ranges tested in the DOE.Red area represents points of failureGreen area represents points of success.,Oval = full design space represented by equation Rectangle represent rangesSimple, but a portion of the design space is not utilizedCould use other rectangles within ovalExact choice of above options can be driven by business factors,For purposes of this case study, an acceptable design space based on ranges was chosen,Seed wt%, ICH, November 2010,Options for Expanding a Design Space,Why expand a Design Space?Business drivers can change, resulting in a different optimum operating spaceWhen is DS Expansion possible?Case A: When the original design space was artificially constrained for simplicityCase B: When some edges of the design space are the same as edges of the knowledge space, ICH, November 2010,API Crystallization: Design Space & Control Strategy,Control Strategy should address:Parameter controls Distillative solvent switch achieves target water contentCrystallization parameters are within the design space TestingAPI feed solution tested for water contentFinal 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, ICH, November 2010,Design Space / Control StrategyParameter controls & Testing,Particle size will be tested in this example, since the result is includedin the mathematical model used for dissolution.,Example from Case Study, ICH, November 2010,Drug Product,Immediate release tablet containing 30 mg AmokinolRationale for formulation composition and process selection providedIn vitro-in vivo correlation (IVIVC) determinationCorrelation shown between pharmacokinetic data and dissolution resultsRobust dissolution measurement neededFor a low solubility drug, close monitoring is important, ICH, November 2010,Drug Product Direct Compression Manufacturing Process,Focus of Story,Example from Case Study,Lubrication, ICH, November 2010,Initial Quality Risk Assessment,Impact of Formulation and Process unit operations on Tablet CQAs assessed using prior knowledgeAlso consider the impact of excipient characteristics on the CQAs,Example from Case Study, ICH, November 2010,Drug Product CQA Dissolution Summary,Quality risk assessmentHigh impact risk for API particle size, filler, lubrication and compressionFillers selected based on experimental work to confirm compatibility with Amokinol and acceptable compression and product dissolution characteristicsAPI particle size affects both bioavailability & dissolutionMultivariate DOE to determine factors that affect dissolution and extent of their impactPredictive mathematical model generatedConfirmed by comparison of results from model vs. actual dissolution testingPossible graphical representations of this design space, ICH, November 2010,Predictive Model for DissolutionA mathematical representation of the design space,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, ICH, November 2010,Dissolution: Control Strategy,Controls of input material CQAsAPI particle sizeControl of crystallisation stepMagnesium stearate specific surface areaSpecification for incoming materialControls of process parameter CPPsLubrication step blending time within design spaceCompression force (set for tablet hardness) within design spaceTablet press force-feedback control systemPrediction mathematical modelUse in place of dissolution testing of finished drug productPotentially allows process to be adjusted for variation (e.g. in API particle size) and still assure dissolution performance, ICH, November 2010,Drug Product CQA -Assay & Content Uniformity Summary,Quality risk assessmentPotential impact for API particle size, moisture control, blending, and lubricationMoisture will be controlled in manufacturing environmentConsider possible control strategy approachesExperimental plan to develop design space using input material and process factorsIn-process monitoringAssay assured by weight control of tablets made from uniform powder blend with acceptable API content by HPLCBlend homogeneity by on-line NIR to determine blending endpoint, includes feedback loopAPI assay in blend tested by HPLCTablet weight by automatic weight control with feedback loop, ICH, November 2010,Blending Process Control Options,Decision on conventional vs. RTR testing,Example from Case Study, ICH, November 2010,Process Control Option 2 Blend uniformity monitored using a process analyser,On-line NIR spectrometer used to confirm scale up of blendingBlending operation complete when mean spectral std. dev. reaches plateau regionPlateau may be detected using statistical test or rulesFeedback control to turn off blenderCompany verifies blend does not segregate downstreamAssays tablets to confirm uniformityConducts studies to try to segregate API,Data analysis model will be providedPlan for updating of model available,Acknowledgement: adapted from ISPE PQLI Team,Example from Case Study, ICH, November 2010,Conventional automated control of Tablet Weight using feedback loop:Sample weights fed into weight control equipment which sends signal to filling mechanism on tablet machine to adjust fill volume and therefore tablet weight.,Tablet Weight Control in Compression Operation, ICH, November 2010,Batch Release Strategy,Finished product not tested for assay, CU and dissolution Input materials meet specifications and are testedAPI particle size distributionMagnesium stearate specific surface areaAssay calculationVerify (API assay of blend by HPLC) X (tablet weight)Tablet weight by automatic weight control (feedback loop), %RSD of 10 tabletsContent UniformityOn-line NIR criteria met for end of blending (blend homogeneity)Tablet weight control results checkedDissolutionPredictive model using input and process parameters calculates for each batch that dissolution meets acceptance criteriaInput and process parameters used are within the filed design spaceCompression force is monitored for tablet hardnessWater content NMT 3% in finished product (not covered in this case study), ICH, November 2010,Drug Product Specifications,Use for stability, regulatory testing, site change, whenever RTR testing is not possibleInput materials meet specifications and are testedAPI PSDMagnesium stearate specific surface areaAssay 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, 2% RSD for weightsContent Uniformity (drug product acceptance criteria meets compendia)On-line NIR criteria met for end of blending (blend homogeneity)Tablet weight control results checkedDissolution (drug product acceptance criteria min 85% in 30 minutes)Predictive model using input and process parameters for each batch calculates whether dissolution meets acceptance criteriaInput and process parameters are all within the filed design spaceCompression force is controlled for tablet hardnessWater content (drug prod

温馨提示

  • 1. 本站所有资源如无特殊说明,都需要本地电脑安装OFFICE2007和PDF阅读器。图纸软件为CAD,CAXA,PROE,UG,SolidWorks等.压缩文件请下载最新的WinRAR软件解压。
  • 2. 本站的文档不包含任何第三方提供的附件图纸等,如果需要附件,请联系上传者。文件的所有权益归上传用户所有。
  • 3. 本站RAR压缩包中若带图纸,网页内容里面会有图纸预览,若没有图纸预览就没有图纸。
  • 4. 未经权益所有人同意不得将文件中的内容挪作商业或盈利用途。
  • 5. 人人文库网仅提供信息存储空间,仅对用户上传内容的表现方式做保护处理,对用户上传分享的文档内容本身不做任何修改或编辑,并不能对任何下载内容负责。
  • 6. 下载文件中如有侵权或不适当内容,请与我们联系,我们立即纠正。
  • 7. 本站不保证下载资源的准确性、安全性和完整性, 同时也不承担用户因使用这些下载资源对自己和他人造成任何形式的伤害或损失。

评论

0/150

提交评论