4-3 documentation quality

Upload: yogesh-n-lala

Post on 04-Apr-2018

225 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/29/2019 4-3 Documentation Quality

    1/55

    Jakarta, October 2009

    Requirements on documentation of API and FPPquality and evaluation process

    Presented byRutendo Kuwana

    Prequalification of Medicines Programme

    QSM / EMP / HSS

  • 7/29/2019 4-3 Documentation Quality

    2/55

    Jakarta, October 2009

    WHO Reference text for Multisource

    (Generic) products / Definitions

    Active Pharmaceutical Ingredient (API)

    A substance or compound that is intended to be used in the manufacture of a

    pharmaceutical product as a therapeutically active compound (ingredient)

    Pharmaceutical Product

    Any preparation for human or veterinary use that is intended to modify or

    explore physiological systems or pathological states for the benefit of therecipient.

    Finished Pharmaceutical Product (FPP)

    A product that has undergone all stages of production, including packaging in

    its final container and labelling.

  • 7/29/2019 4-3 Documentation Quality

    3/55

    Jakarta, October 2009

    Reference texts

    Prequalification quality guidelines for dossier submission

    Guideline on submission of documentation for prequalification of multi-

    source (Generic) Finished Pharmaceutical Products (FPPs) used intreatment of HIV/AIDS, Malaria and Tuberculosis (Main Generic guide with8 annexes) [under revision]

    Supplement 1 : Dissolution testing

    Supplement 2 : Extension of the WHO list of stable APIs (not easilydegradable)

    Guidelines for registration of fixed-dose combination medicinal products

    Guideline on Active Pharmaceutical Ingredient Master File (APIMF)Procedure

    Guidance on variations to a prequalified dossier

    ICH notes for guidance (When WHO or PQ guidelines silent)

    Prequalification of Generic products approved by Stringent RegulatoryAuthorities (SRAs) NEW

    Requalification Draft not finalised and not yet adopted

  • 7/29/2019 4-3 Documentation Quality

    4/55

    Jakarta, October 2009

    Guidelines for Product dossier /Quality

    Main Generic guide

    Guideline on Submission of Documentation for Prequalification of

    Multisource (Generic) Finished Pharmaceutical Products (FPPs) Used inthe Treatment of HIV/AIDS, Malaria and Tuberculosis [under revision]

    Annex 1 - Model Certificate of a Pharmaceutical Product

    Annex 2 - Model Batch Certificate of Pharmaceutical Product

    Annex 3 - Model Stability Report of Active Pharmaceutical Ingredient (API)

    Annex 4 - Model Stability Report of Capsules/Tablets

    Annex 5 - Suggested Structure of the Summary of Product Characteristics(SmPC)

    Annex 6 - Suggested Structure of the Package Information Leaflet (PIL)

    Annex 7 - Presentation of Bioequivalence Trial Information (BTIF)

    Annex 8 - Presentation of Pharmaceutical Quality Information (PQIF)

    http://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFPPs_08_2005_ANNEX1.pdfhttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFpps_08_2005_ANNEX2.pdfhttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFPPs_08_2005_ANNEX3.pdfhttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFPPs_08_2005_ANNEX4.pdfhttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFPPs_08_2005_ANNEX5.pdfhttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFPPs_08_2005_ANNEX6.pdfhttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFPPs_08_2005_ANNEX7.dochttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFPPs_08_2005_ANNEX8.dochttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFPPs_08_2005_ANNEX8.dochttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFPPs_08_2005_ANNEX8.dochttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFPPs_08_2005_ANNEX7.dochttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFPPs_08_2005_ANNEX7.dochttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFPPs_08_2005_ANNEX6.pdfhttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFPPs_08_2005_ANNEX6.pdfhttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFPPs_08_2005_ANNEX5.pdfhttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFPPs_08_2005_ANNEX5.pdfhttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFPPs_08_2005_ANNEX4.pdfhttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFPPs_08_2005_ANNEX4.pdfhttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFPPs_08_2005_ANNEX3.pdfhttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFPPs_08_2005_ANNEX3.pdfhttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFpps_08_2005_ANNEX2.pdfhttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFpps_08_2005_ANNEX2.pdfhttp://healthtech.who.int/pq/info_applicants/Guidelines/GuideGenericSubmitDocFPPs_08_2005_ANNEX1.pdf
  • 7/29/2019 4-3 Documentation Quality

    5/55

    Jakarta, October 2009

    Assessment Process - Quality

    Submission of application

    PQIF, Dossier, BTIF

    Screening (Internal)

    Acceptance

    Pre Assessment Screening

    Assessment

    Report

    Additional data

    Prequalification LoPQ

    Requalification

  • 7/29/2019 4-3 Documentation Quality

    6/55

    Jakarta, October 2009

    Quality dossier / Section 1

    Information on the FPP

    1.1. Details of the Product

    - Name, dosage form and strength of the product

    - Approved generic name (INN)

    - Visual description of the FPP

    - Visual description of the packaging

    1.2. Samples to be provided (for visual examination of assessors and

    comparison with the SPC and PIL)

    1.3. Regulatory situation in Member States / list countries

    - Countries where a MA has been issued

    - Countries where a MA has been withdrawn

    - Countries where a Marketing Application has been rejected, deferred

  • 7/29/2019 4-3 Documentation Quality

    7/55

    Jakarta, October 2009

    Quality dossier / Section 2

    Active Pharmaceutical Ingredient

    (API)

  • 7/29/2019 4-3 Documentation Quality

    8/55

    Jakarta, October 2009

    Quality dossier / Section 2

    Active Pharmaceutical Ingredient (API)

    Scientific data on the API can be submitted using

    following ways and order of preference

    A valid Certificate of Suitability(CoS) or CEP, latest version, with

    all its annexes issued by EDQM

    AnAPIMF(Active Pharmaceutical Ingredient Master File) submittedby the API manufacturer, containing the whole information requested

    in section 2 and presented in CTD format (see APIMF guideline)

    Completesubmission ofdata requested in Section 2

  • 7/29/2019 4-3 Documentation Quality

    9/55

    Jakarta, October 2009

    Quality dossier / Section 2

    of PQIF - Active Pharmaceutical Ingredient (API)

    2.1. Nomenclature

    2.2. Properties of the API

    2.3. Site(s) of manufacture

    2.4. Route(s) of synthesis

    2.5. Specifications

    2.6. Container- closure system

    2.7. Stability testing

  • 7/29/2019 4-3 Documentation Quality

    10/55

    Jakarta, October 2009

    CTD Structure of API Sections

    2.3.S DRUG SUBSTANCE

    2.3.S.1 General Information

    2.3.S.2 Manufacture

    2.3.S.3 Characterisation

    2.3.S.4 Control of Drug Substance

    2.3.S.5 Reference Standards or Materials

    2.3.S.6 Container Closure System

    2.3.S.7 Stability

  • 7/29/2019 4-3 Documentation Quality

    11/55

    Jakarta, October 2009

    2.3.S DRUG SUBSTANCE

    2.3.S.1 General Information

    2.3.S.1.1 Nomenclature

    Include INN, compendial name, chemical name(s),company/laboratory code, other non-proprietary names eg USAN,JAN, BAN and Chemical Abstracts Service (CAS) registry number.

    2.3.S.1.2 Structure

    Include structural formula with relative and absolutestereochemistry, molecular formula, and the relative molecularmass.

    2.3.S.1.3 General Properties

    Physicochemical and other relevant properties of the API.

  • 7/29/2019 4-3 Documentation Quality

    12/55

    Jakarta, October 2009

    2.3.S.2 Manufacture

    Information on the manufacturer;

    A brief description of the manufacturing process

    (including, for example, starting materials, reagents,

    solvents, critical steps, and reprocessing) and the

    controls intended to result in the routine and consistentproduction of API.

    A flow diagram;

    A description of the Source and Starting Material and rawmaterials used in the manufacture of the API;

  • 7/29/2019 4-3 Documentation Quality

    13/55

    Jakarta, October 2009

    2.3.S.2 Manufacture Contd

    A discussion of the selection and justification of criticalmanufacturing steps, process controls, and acceptancecriteria. Discuss critical process intermediates;

    A description of process validation and/or evaluation.

    A brief summary of major manufacturing changes madethroughout development and conclusions from theassessment used to evaluate product consistency. TheQOS should cross-refer to the non-clinical and clinical

    studies that used batches affected by thesemanufacturing changes.

  • 7/29/2019 4-3 Documentation Quality

    14/55

    Jakarta, October 2009

    2.3.S.3 Characterisation

    A summary of the interpretation of evidence of structureand isomerism.

    When an API is chiral, it should be specified whether

    specific stereoisomers or a mixture of stereoisomers have

    been used in the nonclinical and clinical studies, and

    information should be given as to the stereoisomer of the

    API that is to be used in the final product intended for

    marketing.

  • 7/29/2019 4-3 Documentation Quality

    15/55

    Jakarta, October 2009

    2.3.S.3 Characterisation Contd:

    Impurities

    Summary of the data on potential and actual impurities arising fromthe synthesis, manufacture and/or degradation, and should

    summarise the basis for setting the acceptance criteria for

    individual and total impurities.

    Summary of the impurity levels in batches of the API used in the

    non-clinical studies, in the clinical trials, and in typical batches

    manufactured by the proposed commercial process. The QOS

    should state how the proposed impurity limits are qualified.

    A tabulated summary, with graphical representation, where

    appropriate should be included.

  • 7/29/2019 4-3 Documentation Quality

    16/55

    Jakarta, October 2009

    2.3.S.4 Control of Drug Substance

    A summary of the specification(s), the analyticalprocedures, and validation should be included.

    A tabulated summary of the batch analyses.

  • 7/29/2019 4-3 Documentation Quality

    17/55

    Jakarta, October 2009

    2.3.S.5 Reference Standards or Materials

    Information on primary and working standards of the APIand specified impurities.

  • 7/29/2019 4-3 Documentation Quality

    18/55

    Jakarta, October 2009

    2.3.S.6 Container Closure System

    A brief description and discussion of the primary andsecondary packaging components.

  • 7/29/2019 4-3 Documentation Quality

    19/55

    Jakarta, October 2009

    2.3.S.7 Stability

    This section should include a summary of the studiesundertaken (conditions, batches, analytical procedures)

    and a brief discussion of the results and conclusions, the

    proposed storage conditions, retest date or shelf-life.

    The post-approval stability protocol should be included.

  • 7/29/2019 4-3 Documentation Quality

    20/55

    Jakarta, October 2009

    Quality dossier / Section 2

    Active Pharmaceutical Ingredient (API)

    PQ dossier section 2CTD2.1 Nomenclature

    2.2 Properties of API (s)

    S.1 General Information- Nomenclature

    - Structure

    - General Properties

    2.3 Site(s) of Manufacture

    2.4 Route(s) of synthesis- API not described in pharmacopoeia

    - Specifications of raw materials and intermediates used in

    synthesis

    - API described in a pharmacopoeia

    S.2 Manufacture- Manufacturer- Description of manufacturing process

    - Control of materials- Control of critical steps and intermediates

    - Process validation

    - Manufacturing process development

    S.3 Characterisation- Elucidation of structure

    - Impurities

    2.5 SpecificationsS.4 Control of Drug Substance

    S.5 Reference Standards or Materials

    2.6 Container Closure System

    2.7 Stability testing

    S.6 Container Closure System

    S.7 Stability testing

  • 7/29/2019 4-3 Documentation Quality

    21/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product

    (FPP)

  • 7/29/2019 4-3 Documentation Quality

    22/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)3.1. Manufacturing and marketing authorization

    3.2. Pharmaceutical development

    3.3. Formulation

    3.4. Sites of manufacture

    3.5. Manufacturing process

    3.6. Manufacturing process controls of Critical steps and intermediates

    3.7. Process validation and Evaluation

    3.8. Specifications for excipients

    3.9. Control of the FPP

    3.10. Container/closure system (s) and other packaging

    3.11. Stability testing

  • 7/29/2019 4-3 Documentation Quality

    23/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.12. Container labelling

    3.13. Product information for health professionals

    3.14. Patient information and package leaflet

    3.15. Justification for any differences to the product in the country

    ORcountries issuing the submitted WHO-type certificate(s)

  • 7/29/2019 4-3 Documentation Quality

    24/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.1. Manufacturing and Marketing Authorization

    - Valid manufacturing authorization for pharmaceuticalproduction including the pharmaceutical form applied for

    - Marketing authorization to demonstrate the product isregistered / licensed in accordance with national requirements

    3.2. Pharmaceutical development

    The aim is to build a quality product by design.

  • 7/29/2019 4-3 Documentation Quality

    25/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.2. Pharmaceutical development

    The section should contain information on the development studiesconducted to establish that

    the dosage form,

    the formulation, the manufacturing process,

    the container closure system,

    microbiological attributes and

    storage and usage instructions

    are appropriate for the purpose specified in the dossier.

  • 7/29/2019 4-3 Documentation Quality

    26/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.2. Pharmaceutical development (pre-formulation)

    Physico-chemical characteristics of the APIs

    solubility (composition)

    water content (stability)

    hygroscopicity (stability)

    particle size (solubility, bioavailability, suspension properties,

    stability )

    polymorphism (solubility, bioavailability, stability)

    Data obtained from literature : Books, Journals, International Pharmaceutical

    Abstracts, Chemical Abstracts, Analytical Abstracts, Internet

    Experimental data (if necessary)

  • 7/29/2019 4-3 Documentation Quality

    27/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.2. Pharmaceutical development (choice of excipients)

    Intended function of each excipient

    Criteria compatibility of excipients with API(s),

    characteristics of the excipients (water content, particle size, flowability,density, rheological behavior)

    Particularly : other non active constituents (lowest acceptableconcentration to be chosen e.g. concentration of parabens aspreservatives)

    Experimental data needed.

  • 7/29/2019 4-3 Documentation Quality

    28/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.2. Pharmaceutical development (choice of the manufacturing

    process)

    Parameters : characteristics of the APIs, dosage form, composition. .

    Rational behind the choice (e.g. why a non over kill process as a sterilisationprocess instead of terminal sterilisation in final container)

    Justification of the overage (if any)

    Identification of the critical steps

    In Process Control (IPC)

    Selection and optimisation of manufacturing process

    Q li d i / S i 3

  • 7/29/2019 4-3 Documentation Quality

    29/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.2. Pharmaceutical development (dissolution testing)

    To study dissolution operating conditions (media, pH, rotation, )

    To develop a discriminatory dissolution method

    Comparative dissolution testing is a tool, mandatory in developmentpharmaceutics section of the dossier in PQ, See Supplement 1

    Help in selection of the formulation

    - compare formulation(s) with innovator product,

    - a basic strategy in development to maximize thechances of bioequivalence

    Comparison of pivotal batches to commercial batches/ post-approval changes

    Setting of dissolution specifications

    Q li d i / S i 3

  • 7/29/2019 4-3 Documentation Quality

    30/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)3.2. Pharmaceutical development (comparative dissolution testing)

    1. Three media - 900 ml or less - all at 37C Buffer pH 1.2 or0.1M HCl

    Buffer pH 4.5

    Buffer pH 6.8

    Water may be usedadditionally (not instead of)

    2. Paddle at 50 orbasket at 100 rpm

    3. Twelve units of each product in all 3 media

    4. Dissolution samples collected at short intervals, e.g. 10, 15, 20, 30, 45 and 60 minutes Analyse samples for all APIs, when applicable

    5. Calculate similarity factor f2

  • 7/29/2019 4-3 Documentation Quality

    31/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.2. Pharmaceutical development (details of batches

    studied)

    Provide a summary of development of the FPP from pre-

    formulation to production scale.

    Provide a comparison of formulas (tabulated form) of: bio-batche(s) (clinical / bioequivalence),

    development batches,

    stability batches,

    batches for validation/production

  • 7/29/2019 4-3 Documentation Quality

    32/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.2. Pharmaceutical development (choice of formulation

    and compatibility)

    Compatibility of APIs with the excipients

    Compatibility of APIs between each other in case of fixed dose

    combinations

  • 7/29/2019 4-3 Documentation Quality

    33/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.3. Formulation

    Formula in tabulated form for :

    Administration unit (e.g. one tablet),

    Typical batch

    - Precise any overage,

    - Precise quantity adjustment of the API,

    - Precise q.s. for excipient.

    Excipients :

    State function (e.g. lubricant, disintegrant),

    Precise technical grade (e.g. micronised, purified water),

    describe also those removed during process (e.g. water),

    Describe also those not always added (e.g. acid & alkali for pH adjustment,

    Capsule shells, inked imprints on dosage form, Also gas (inert atmosphere).

    Q / S 3

  • 7/29/2019 4-3 Documentation Quality

    34/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.4. Manufacturing sites

    Name and street address of each facility where any aspect ofmanufacture occurs including production, sterilisation,packaging and quality control include Units and/or Blocks

    Include any alternative manufacturers

    Certificate issued by the Competent DRA according to WHOCertification scheme for each site where a major step ofmanufacturing is performed

    Submit a valid GMP certificate

    Q lit d i / S ti 3

  • 7/29/2019 4-3 Documentation Quality

    35/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.5. Manufacturing process (cont.)

    Flow chart with indication of each step showing where materials

    enter the process. Indication of critical steps and in-process

    controls

    Description of manufacturing/packaging including

    Scale

    Equipment by type (e.g. tumble blender) & working capacity

    Process parameters for steps, (e.g. time, temperature, pH)

    Environmental conditions, e.g. relative humidity for hygroscopic

    FPPs., area class for sterile FPPs

    Steps of the process

    Alternative methods

    Q lit d i / S ti 3

  • 7/29/2019 4-3 Documentation Quality

    36/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.5. Manufacturing process (cont.)

    Proposal for reprocessing justified with data.

    Copy of master formula.

    Batch manufacturing record real batch.

    Sterile products sterilisation steps and/or aseptic procedures.

    Description of in-process tests including plan of sampling and acceptance

    limits.

    Data for 3 full scale batches to support achievement of predetermined

    specifications.

    Q lit d i / S ti 3

  • 7/29/2019 4-3 Documentation Quality

    37/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.6. Manufacturing Process Controls of Critical steps and

    Intermediates

    Identification of critical steps with test methods and justified

    acceptance criteria

    Information on quality of isolated intermediates, test methods

    and justified acceptance criteria to control them

    Q lit d i / S ti 3

  • 7/29/2019 4-3 Documentation Quality

    38/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.7. Process Validation and Evaluation

    Validation mandatory for processes including a critical step

    The aim is to show that critical steps are under control and lead continuously tothe desirable quality

    Examples of critical steps (list non exhaustive) mixing,

    coating,

    granulation,

    emulsification,

    non-standard sterilisation

    Q lit d i / S ti 3

  • 7/29/2019 4-3 Documentation Quality

    39/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.7. Process Validation and Evaluation (details on 3 first production batches)

    Batches. batch number

    . batch size

    . place and date of manufacture

    . batch number of API(s)

    . yield

    . batch purpose (validation, stability, clinical trial )

    Process. equipment

    . process parameters

    . validation protocol.

    Results

    . critical steps

    . in process control

    . finished product

    Quality dossier / Section 3

  • 7/29/2019 4-3 Documentation Quality

    40/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.7. Process Validation and Evaluation

    Concurrent validation carried out during normal production on

    the first 3 production batches

    OR

    For well-established processes

    process data, in-process controls and quality controls on a total

    of 10- 25 batches to present a statistically significant picture

    Q lit d i / S ti 3

  • 7/29/2019 4-3 Documentation Quality

    41/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.7. Process Validation and Evaluation

    If validation data (on production scale batches) are not

    available submit

    validation protocol,

    commitment that validation report will be submitted later for

    evaluation,

    commitment that data will be available in case of inspection,

    commitment that WHO will be informed of any significant

    deviation.

    Q lit d i / S ti 3

  • 7/29/2019 4-3 Documentation Quality

    42/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.7. Process Validation and Evaluation

    Validation protocol should include

    brief description of the process with summary of critical steps and parameters to befollowed during validation,

    specifications of the FPP at release,

    details of analytical procedures and limits,

    sampling plan,

    unifromity of dosage units essential for FDCs,

    proposed timeframe

    Validation report when submitted should includeresults for each batch, certificates of analysis, batch production records,report on unusual findings, modifications, observations and conclusions

    Quality dossier / Section 3

  • 7/29/2019 4-3 Documentation Quality

    43/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)3.8. Specifications for excipients

    - Non pharmacopoeial substances- Details for manufacturing process,- Specifications (description of procedures and acceptance criteria)- Stability data- Cross-reference to non-clinical (toxicological)- clinical data for safety aspects- Certificates of analyses

    - Pharmacopoeial excipients

    Copy of the pharmacopoeial monograph used for control + certificates of analysis

    - For excipients of animal, human, microbial origin

    - TSE (Transmissible Spongiform Encephalopathy) risks and viral safety should beaddressed- TSE CEP preferred

    Permitted colorantsare those allowed in UE, USA and Japan

    Quality dossier / Section 3

  • 7/29/2019 4-3 Documentation Quality

    44/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.9. Control of the FPP

    2 sets of specifications are possible: at release and at the end of shelf-life(list of attributes non exhaustive)

    Description of the FPP /appearance

    Identification of API

    Assay of API: 5% of the label claim at release and 10% at the end of shelf-life

    Degradation products

    Pharmaceutical tests e.g. dissolution, disintegration (where applicable)

    Uniformity of dosage units (mass or content)

    Identification of colorants, identification and assay of anti-oxidants, chemical

    preservatives

    Microbial contamination, Sterility, bacterial endotoxins

    Q lit d i / S ti 3

  • 7/29/2019 4-3 Documentation Quality

    45/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.9. Control of the FPP (cont.)

    Monographs of Ph. Int., USP, BP are acceptable for the FPP +

    complementary tests

    If non-pharmacopoeial FPP, note for guidance Q6A applicable

    Description of all analytical procedures in details if not described in a

    pharmacopoeial monograph

    Validation of analytical methods and/or demonstration of applicability for

    pharmacopoeial methods

    Batch analyses for 3 lots with details of each lot (batch no, size, date of

    manufacture, use of batch)

    Quality dossier / Section 3

  • 7/29/2019 4-3 Documentation Quality

    46/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.10. Container-closure system

    Discussion on the choice of container

    Choice of the materialProtection against light and humiditycompatibility/interaction of materials in contact with dosage formSafety of materials used

    Detailed description of the container

    Specifications of the container with dimensions and drawingsSpecifications of materials in contact with FPPComposition of these materials, compliance with pharmacopoeiaIdentification of components e.g. IR for plastic materials

    Description of the secondary packaging

    Quality dossier / Section 3

  • 7/29/2019 4-3 Documentation Quality

    47/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.11. Stability testing

    The purpose of stability testing is to provide evidence on how the

    quality of a FPP varies with time under the influence of a variety

    of environmental conditions such as temperature, humidity

    and light and to establish a shelf-life for the FPP, to determine

    the storage conditions and the in-use stability.

    Quality dossier / Section 3

  • 7/29/2019 4-3 Documentation Quality

    48/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.11. Stability testing

    Lots included in the study: 1 production batch and 2 of pilot scalemanufactured according to the process described in the dossier3 Pilotscale batches are acceptable(exception for 2nd line TB products: 2 pilot batches)

    Pilot scale batch for solid dosage forms is 10% of production scale or100 000 whichever is greater

    Parameters susceptible to change over storage should be followed:

    . Organoleptic properties

    . Assay of each API: 10% of the label claim possible at the end of shelf-life

    . Assay of degradation products

    . Assay of antioxidants and chemical preservatives, check also for their efficacy

    . Dissolution testing (limits should remain unchanged to release)

    . Microbial contamination, sterility, bacterial endotoxins

    In-use stability data (if applicable)

    Quality dossier / Section 3

  • 7/29/2019 4-3 Documentation Quality

    49/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.11. Stability testing

    Study should be performed in the claimed commercial packaging (container-closure)

    Storage conditions and frequency of testing according to WHO stability guideline inTRS 953

    Minimum stability data to be submitted at time of submission:

    Long term 12 months or 6 months or 3 months (as appropriate according toSupplement 2 to the Main Generic guide and exception for TB 2nd line

    products) 6 months intermediate 30C/65% RH

    6 months accelerated 40C/75% RH

    Unless otherwise justified, 30C / 75% RH is the recommended storage condition forPrequalification

    Definition of "significant change" in WHO stability guide is the same as ICH Q1A (R2)

    Quality dossier / Section 3

  • 7/29/2019 4-3 Documentation Quality

    50/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.11. Stability testing

    Case of products packed in semi-permeable containers foreseen (liquiddosage forms susceptible to loss of solvent or water loss in low relativehumidity condition). The storage condition will be long term ICH 25C / 40%RHOR30C/35% RH and accelerated 40C/25% RH

    Extrapolation of data to accord a longer shelf-life possible according to ICHQ1E + Supplement 2 in condition of commitments

    Supplement 2: tentative 2 year re-test period and /or shelf-life may beaccorded to APIs and corresponding solid forms (tablets and capsules)listed in Supplement 2 based only on 6 months accelerated data and 6months long term data

    Long term stability should anyhow be followed to cover the whole shelf-life

    accorded

    Quality dossier / Section 3

  • 7/29/2019 4-3 Documentation Quality

    51/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.12. Container labellingOuter packaging : Where no outer packaging, on immediatepackaging, e.g. HDPE bottle.

    Labelling should include at least the following :

    The name of the FPP.

    Method of administration.

    A list of API(s) (using INNs if applicable) showing the amount of each present in adosage unit, and a statement of the container, e.g. number of dosage units, weight or

    volume. List of excipients known to be a safety concern for some patients, e.g. lactose, gluten,

    metabisulfites, parabens, ethanol, or tartrazine.

    Instruction on use.

    The batch number assigned by the manufacturer.

    The expiry date in an uncoded form.

    Storage conditions or handling precautions that may be necessary.

    Directions for use, and any warnings or precautions that may be necessary.

    The name and address of the manufacturer, company or person responsible for placingthe product on the market.

    Quality dossier / Section 3

  • 7/29/2019 4-3 Documentation Quality

    52/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.12. Container labelling

    Blisters and stripsshould include, as a minimum, the following

    information

    Name, strength and pharmaceutical form of the FPP

    Name of the manufacturer, company or person

    responsible for placing the product on the market

    The batch number assigned by the manufacturer

    The expiry date in an un-coded form

    Quality dossier / Section 3

  • 7/29/2019 4-3 Documentation Quality

    53/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.13. Product information for Health Professionals

    Summary of product characteristics (SmPC)

    Aimed at medical practitioners and health professionals Changes to SmPC to be approved by WHO

    See Annex 5 of the main generic guide

    Quality dossier / Section 3

  • 7/29/2019 4-3 Documentation Quality

    54/55

    Jakarta, October 2009

    Quality dossier / Section 3

    Finished Pharmaceutical Product (FPP)

    3.14. Patient information and package leaflet

    Copy of the patient information leaflet (PIL)

    In conformance with SmPC

    See Annex 6 of the main Generic guide

  • 7/29/2019 4-3 Documentation Quality

    55/55

    Thank youfor your attention