Finished Pharmaceutical Products (FPPs) (special reference to TB and HIV/AIDS) - PowerPoint PPT Presentation

1 / 50
About This Presentation
Title:

Finished Pharmaceutical Products (FPPs) (special reference to TB and HIV/AIDS)

Description:

(special reference to TB and HIV/AIDS) World Health Organization ... State function (e.g. lubricant, disintegrant) Technical grade (e.g. micronised, purified water) ... – PowerPoint PPT presentation

Number of Views:279
Avg rating:3.0/5.0
Slides: 51
Provided by: puk3
Category:

less

Transcript and Presenter's Notes

Title: Finished Pharmaceutical Products (FPPs) (special reference to TB and HIV/AIDS)


1
Finished Pharmaceutical Products (FPPs)(special
reference to TB and HIV/AIDS)
  • World Health Organization
  • Training Workshop on Pharmaceutical Quality, GMP
    and Bioequivalence
  • Kiev - Ukraine
  • 3 to 7 October 2005

Theo Dekker, D.Sc., consultant to WHO Research
Institute for Industrial Pharmacy North-West
University, Potchefstroom, South
Africa iiftgd_at_puk.ac.za
2
What is an FPP?
  • 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
    the recipient
  • Finished Pharmaceutical Product (FPP)
  • A product that has undergone all stages of
    production, including packaging in its final
    container and labelling
  • Marketing Authorization of Pharmaceutical
    Products with special Reference to Multisource
    (Generic) Products a Manual for a Drug
    Regulatory Authority(WHO, Blue Book)

3
Section 3. Dossier requirements for a Finished
Pharmaceutical Product (FPP)
  • Guideline
  • Guideline on Submission of Documentationfor
    Prequalification of Multi-source (Generic)
    Finished Pharmaceutical Products (FPPs)used in
    the Treatment of HIV/AIDS, Malaria and
    Tuberculosis
  • (Main guideline, PQ website, hand-out)

4
Section 3. FPPs content of dossier 10/50
  • 3.1 Manufacturing and marketing authorization
    10/50
  • 3.2 Pharmaceutical development 10/50
  • 3.3 Formulation 11/50
  • 3.4 Sites of manufacturing 12/5
  • 3.5 Manufacturing process 12/5
  • 3.6 Process controls, critical steps
    intermediates 12/5
  • 3.7 Process validation and evaluation 13/5
  • 3.8 Specifications for excipients 15/50
  • 3.9 Control of the FPP (specifications) 16/50
  • 3.10 Container closure system other packaging
    17/50
  • 3.11 Stability testing (shelf-life) 17/50
    (separate topic)
  • 3.12, 3.13, 3.14 Labelling/SmPC/PIL 23/50
  • Not discussed in session

5
3.2 Pharmaceutical development 10/50
  • The aim of pharmaceutical development is
  • to design a quality product and
  • the manufacturing process to deliver the product
    in a reproducible manner.
  • Quality is built in by design, not tested in
  • ICH Q8

6
3.2 Pharmaceutical developmentDesk research
  • Learn about FPP before development, through
  • Standard works / series / books such as
  • (Analytical) Profiles of Drug Substances and
    Excipients eds Florey / Brittain 31 volumes
  • Especially for older products, such as TB
  • Journals through search facilities such as
  • International Pharmaceutical Abstracts, Chemical
    Abstracts, Analytical Abstracts internet
  • Public assessment reports
  • EPAR WHOPAR
  • Particularly important for ARVs

7
3.2 Pharmaceutical developmentDesk research TB
4FDC tablets (1)
  • This is a problem combination
  • Composition in current Essential Drug List
  • Rifampicin 150 mg
  • Isoniazid 75 mg
  • Pyrazinamide 400 mg
  • Ethambutol 2HCl 275 mg
  • Total API weight 900 mg
  • Typical tablet weight 1.3 g

8
3.2 Pharmaceutical developmentTB 4FDC tablets
a problem mix (2)
  • Rifampicin
  • Oxidation (quinone N-oxide)
  • Protect from air exposure
  • Hydrolysis (3-formylrifamycin 25-desacetyl)
  • Wet granulation / drying a potential problem?
  • Reaction with Isoniazid
  • produces 3-(isonicotinylhydrazinomethyl)rifamycin
    or more commonly known as isonicotinyl hydrazone
  • isonicotinyl hydrazone major decomposition
    product
  • Light sensitive
  • Product to be protected from light exposure

9
3.2 Pharmaceutical developmentTB 4FDC tablets
a problem mix (3)
  • Isoniazid
  • Reacts with aldehydes/reducing sugars
  • Sugar lactose to be avoided in formulation !!
  • 3-Formylrifamycin (from rifampicin)
  • Ethambutol hydrochloride (2HCl)
  • Hygroscopic
  • Absorbs water for reaction in tablets
  • Creates slightly acidic conditions
  • pH of 2 w/v solution 3.7-4.0 (BP)
  • The acidic conditions enhance reaction between
    rifampicin and isoniazid reaction (isonicotinyl
    hydrazone formation)

10
3.2 Pharmaceutical developmentTB 4FDC tablets
a problem mix (4)
  • Isonicotinyl hydrazone (3-(isonicotinylhydrazinome
    thyl)rifamycin)
  • This is major decomposition product in tablets
    containing rifampicin and isoniazid
  • Series of articles by dr. S. Singh et al.
    (NIPER), e.g.
  • S. Singh, T. T. Mariappan, N. Sharda, S. Kumar
    A. K. Chakraborti. The reason for an increase in
    decomposition of rifampicin in the presence of
    isoniazid under acid conditions. Pharm.
    Pharmacol. Commun., 6, 405-410 (2000)
  • The pathway for the formulation of the hydrazone
    is discussed in the above publication

11
4FDC-TB tabletsexposed to 40C/75RH for one week
  • Two products. Bleeding may start after more
    exposure (in-house) ethambutol hygroscopic
  • Indicates problem of moisture
  • Control on left Control on left

12
4FDC-TB tabletspreventative/protective measures
  • Formulation - no sugar/lactose (isoniazid)
  • Separate granulation of rifampicin isoniazid
    (limit contact)
  • Rifampicin as powder (not granulate)?
  • Prevent oxidation hydrolysis
  • Low water content of tablet (USP 3.0)
  • Protect product from moisture and oxygen
  • Film coating
  • Non-permeable packaging
  • Do not remove from primary packaging
  • Avoid repackaging
  • Light protection
  • Enteric coated tablets (delayed release)?
    clinical studies
  • Differential formulation, e.g. delayed release
    immediate release in one tablet ?? clinical
    studies

13
3.2. Dossier requirementsPharmaceutical
development
  • Section 3.2 should contain information on the
    development studies conducted to establish that
  • the dosage form,
  • the formulation,
  • manufacturing process,
  • container closure system,
  • microbiological attributes and
  • storage and usage instructions
  • are appropriate for the purpose specified in the
    application.
  • Also consult ICH Q8 Pharmaceutical Development

14
3.2.1 Company research and development (RD) 10/50
  • Aspects important for development of tablets
  • a) Properties of the API of importance to the for
    FPP manufacturing and performance to be
    discussed, supported by data, such as
  • Stability for instance against
  • heat, oxidation by air and hydrolysis
  • Solubility
  • Hygroscopicity
  • Crystal form and particle size
  • especially for practically insoluble APIs

15
3.2.1 Company RD (cont.)
  • Choice of excipients, e.g.
  • Compatibility with API(s)
  • Intended functions and concentrations in product
  • Safety aspects, e.g. TSE risk, to be addressed
  • For fixed-dose combination (FDC) products
  • Compatibility of APIs with each other
  • Consult the FDC guidelines of WHO and Botswana
    Principles on website

16
3.2.1 Company RD (cont.)
  • Comparative dissolution testing (separate topic)
  • Assist in selection of the formulation
  • Compare formulation(s) with innovator product
  • This should be a basic strategy in development to
    maximize the chances of bioequivalence
  • Comparison of pivotal batches
  • Setting of dissolution specifications
  • e) Explain
  • selection and
  • optimisation
  • of manufacturing process critical aspects

17
3.2.1 Company RD (cont.)
  • f) Overages should be justified
  • Packaging should ensure stability of FPP
  • Prospective validation at development phase
  • critical situations
  • unsatisfactory processes to be rectified
  • See also ICH Q8 guideline

18
3.2.1 Company RD (cont.)
  • i) Comparison of formulas (tabulated form) of
  • bio-batche(s) (clinical / bioequivalence)
  • development batches
  • stability batches
  • batches for validation / production
  • Provide brief summary of development of the FPP
    from pre-formulation to production scale

strict requirement
19
3.2.1 Information from literature 11/50
  • Supportive data from literature can be included
  • Strongly recommended
  • Scientifically sound do not reinvent the wheel
  • Examples
  • Examples given at this workshop in other
    presentations
  • 4FDC TB tablets incompatibilities (earlier)
  • Indinavir sulfate
  • Excellent analysis of published data for
    development purposes Dr. J. Pogány Research
    Development, WHO workshop, 28 February 2005,
    Shanghai (PQ website)
  • Solid state properties of rifampicin (next slides)

20
3.2.1 Information from literatureRifampicin
solid state properties (1)
  • Rifampicin exist is 3 solid state forms
  • Polymorph I
  • Polymorph II
  • Amorphous form
  • Commercial material contains
  • Polymorph II (predominantly)
  • Mixture of polymorph II and amorphous form
  • Five commercial samples (A to E) tested
  • Sample A Form II
  • Sample B Form II
  • Sample C Form II amorph
  • Sample D Form II amorph
  • Sample E Form II

21
3.2.1 Information from literature Rifampicin -
SEM photos
  • Sample A Sample D
  • Form II Form II amorph

22
3.2.1 Information from literatureRifampicin
-XRPDs
  • Top Sample A (Form II) sharp signals
  • Middle Sample C (Form II amorph) intensity
    drop
  • Bottom Amorphous form - no pattern

23
3.2.1 Information from literatureRifampicin
powder dissolution (1)
  • Medium 0.1 M hydrochloric acid
  • Profiles of all samples are similar
  • Dissolves immediately in 0.1 M hydrochloric acid

24
3.2.1 Information from literatureRifampicin
powder dissolution (2)
  • Medium Phosphate buffer pH 7.4
  • Profiles A, B E are similar (f2 50)
  • Profiles C D are similar (f2 50) -
    dissolution incomplete
  • Profiles A, B, E dissimilar from profiles C,D (f2
    lt 50)

A, B, E(form II)
C, DForm II Amorph
25
3.2.1 Information from literatureRifampicin
powder dissolution (3)
  • Medium Water
  • Profiles A, B E are similar (f2 50)
  • Profiles C D are similar (f2 50) -
    dissolution incomplete
  • Profiles A, B, E dissimilar from profiles C,D (f2
    lt 50)

A, B, E(form II)
C, D(form II amorph)
26
3.2.1 Information from literatureRifampicin -
solid state conclusions
  • Solid state forms can be identified with XRPD
    SEM
  • Presence of amorphous form slows down dissolution
    of raw material powder at higher pH (f2 test)
  • Incomplete dissolution after 65 minutes !!
  • Product may fail USP tolerance at pH 6.8 (75 in
    45 min.)
  • Reason Agglomeration / wettability
  • Comparative powder dissolution powerful tool for
  • selection of API manufacturer and
  • even in-house specification
  • Reference
  • S. Q. Henwood et al. Solubility and dissolution
    properties of generic rifampicin raw materials.
    Drug Dev. Ind. Pharm. 26, 403-408 (2000) (RIIP)

27
3.3 Formulation 11/50
  • Formula in tabulated form for
  • Administration unit (e.g. one tablet)
  • Typical batch
  • Excipients
  • State function (e.g. lubricant, disintegrant)
  • Technical grade (e.g. micronised, purified water)
  • Also those removed during process (e.g. water)
  • Also those not always added (e.g. acid alkali)
  • Capsule shells, inked imprints on dosage form
  • Justify any overages

28
3.3 FormulationExample
  • ? Removed during process (not in total mass)

Ingredient Quantity per tablet (mg) Quantity per batch (kg) Purpose
Isoniazid 300.00 600.00 Active
---------------- ------(90.00) ------(180.00) --------------
Mg Stearate 2.00 4.00 Lubricant
Pur. Water? 60.0 120.00 Solvent
Total 450.00 900.00
2 000 000 tablets 2 000 000 tablets
29
3.5 Manufacturing process 12/50
  • Flow diagram
  • Indicate critical steps 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, temp, pH
  • Environmental conditions, e.g. rel. humidity for
    hygroscopic FPPs.

30
3.5 Manufacturing process (cont.)
  1. Proposal for reprocessing justified with data
  2. Copy of master formula
  3. Batch manufacturing record real batch
  4. Sterile products sterilisation steps /or
    aseptic procedures
  5. Description of in-process tests
  6. Data for 3 full scale batches to support
    achievement of predetermined specifications

31
3.6 Manufacturing process controls ofcritical
steps and intermediates 12/50
  • Critical steps
  • Acceptance criteria (justified)
  • Test methods (cross reference acceptable)
  • Intermediates isolated during process,
  • for instance tablets cores in film-coated tablet
    production
  • Acceptance criteria (justified if not compendial)
  • Test methods (cross reference acceptable)

32
3.7 Process validation evaluation 13/50
  • Differentiate between the following generics
  • 3.7.1 New FPPs (new for manufacturer, not
    market)
  • FPPs that have been newly developed by the
    manufacturer, though it will be a generic
  • Full validation required
  • 3.7.2 Established FPPs
  • The manufacturer has manufactured marketed this
    FPP for quite some time and now wishes to
    prequalify the FPP
  • 10 recent consecutive batches
  • result/trend/statistical analysis discussion
  • Rejected batches excluded - submit failure
    investigation

33
3.7.1 Validation new product 13/50
  • Purpose To demonstrate validity of process
  • Submit concurrent validation
  • If not yet available
  • Submit validation protocol (details in dossier)
  • Commitment that once data has been generated on
    production scale batches
  • the data will be available for verification
    during inspection
  • the validation report will be submitted for
    evaluation
  • WHO will be informed of any significant
    deviations from the expected and proposed changes
    need prior approval by WHO by way of variation

34
3.7.1 Validation new productConcurrent /
prospective validation (1)
  • Concurrent validation
  • Carried out during normal production
  • First 3 production batches (prospective
    validation)
  • In-process controls are set on outcome
  • Extensive sampling and testing during process,
    for example (tablets)
  • planned sampling on mixing / granulation stages
    for content uniformity (low-dose FPPs FDCs !)
  • A large number tablets for mass and/or content
    uniformity, hardness, friability and even
    dissolution
  • Sampled according to plan during process
  • Statistical analysis of results with conclusions
  • To be within acceptance criteria

35
3.7.1 Validation new productConcurrent /
prospective validation (2)
  • Parenteral products, aseptically filled (if
    terminal sterilization is not possible)
  • Filling ampoules with culture media, then
  • Incubation and control of microbial growth
  • Level of contamination 0.1 (previously
    0.3)
  • Challenge experiments to determine
  • robustness of process
  • affect of material variations, such as particle
    size
  • Can be carried out on experimental batches
  • For instance stability of granulate over time
  • Effect in case of unplanned stoppage

36
3.7.1 Validation new productConcurrent /
prospective validation (4)
  • Laboratory scale batches (small size),
  • To support e.g. formulation and packaging
    development
  • Pilot batches
  • Used e.g. in stability and safety/efficacy
    studies
  • Size for oral solid dosage forms the largest of
  • 10 of production scale or 100,000 units
  • Productions scale
  • For full validation and stability studies
  • Scale-up / scale down after prequalification
  • Up to10-fold compared to the original batch size
    (minor)
  • Consult guideline on variations revalidation
    required

37
3.9 Control of FPP 16/50
  • Four subsections
  • 3.9.1 Specifications
  • 3.9.2 Analytical procedures
  • 3.9.3 Validation of analytical procedures
  • 3.9.4 Batch analysis (against full set of
    specifications)
  • Three or more batches
  • Full information on the batches, e.g.
  • Batch number and size
  • Date/place of manufacture and QC testing
  • Purpose of batches
  • Batch number of API
  • QA certified

38
3.9.1 Specifications for the FPP 16/50
  • Specifications are one part of a total control
    strategy for the FPP designed to ensure product
    quality and consistency (ICH Q6A).
  • Others include sound development studies and
    adherence to GMP e.g., suitable facilities, a
    validated manufacturing process, in-process
    testing, stability testing, API testing, etc.
  • Product specifications (as in pharmacopoeia) or
    split into
  • Release specifications and
  • Shelf-life specifications (may differ if
    justified)

39
3.9.1 FPP specifications (cont.)
  • Important reading for setting specifications
  • ICH guideline Q6A (also good for generics)
  • Specifications Test Procedures and Acceptance
    Criteria for New Drug Substances and New Drug
    Products Chemical Substances.
  • Specifications based on compendial monographs
  • Additional product related specifications, e.g.
  • Those standard for the type of dosage form (e.g.
    friability, tablet hardness, mass uniformity,
    viscosity)
  • ID of colorants (skip testing?)
  • microbial limits (skip testing?)
  • ID and assay of preservatives
  • Limits may be tighter than in monograph

40
3.9.1 FPP specificationsTypical parameters (2)
  • Appearance
  • Identification of the following in FPP
  • APIs
  • Colorants (skip testing possible)
  • Preservatives
  • Physical tests appropriate to dosage form e.g.
  • LOD, friability, hardness (tabs), relative
    density
  • Uniformity of dosage units (mass / content)
  • Pharmaceutical tests, e.g.
  • dissolution
  • Each API in FDC products

41
3.9.1 FPP specificationsTypical parameters (2)
  • Purity tests
  • Degradation products (related substances)
  • Special attention to API-API degradation products
  • Residual solvents (solvents used in process)
  • Microbial count / sterility / bacterial
    endotoxins
  • Content of APIs in FPP (assay)
  • Limits 95.0 105.0, unless justified
  • Content of preservatives
  • Limits 90.0 110.0, generally acceptable

42
3.9.1 FPP specificationsExample for uncoated
tablets (1)
Attribute Release limits Stability limits
Appearance Full description Same as release
Identification At least 1 method Not required for stability studies. Not regarded as variables for product.
Dimensions Diameter, etc Not required for stability studies. Not regarded as variables for product.
Average mass w.r.t. theoretical Not required for stability studies. Not regarded as variables for product.
Mass uniformity Ph.Eur/USP/Int.Ph Not required for stability studies. Not regarded as variables for product.
Tablet hardness product specific Same as release
43
3.9.1 FPP specificationsExample for uncoated
tablets (cont.)
Attribute Release limits Stability limits
Friability 1 (normally) Same as release
Dissolution Set per product Same as release
Disintegration Not required if dissolution is done Not required if dissolution is done
Rel. substances (degradants) Only if formed during production Required. Limits to one decimal
Assay (content) 95.0-105.0, unless justified May be 90.0-105.0, if justified
Microbial limits Skip-testing end of shelf
Tests not necessary at release if done in-process Tests not necessary at release if done in-process Tests not necessary at release if done in-process
44
3.9.3 Validation analytical methods 17/50
  • Non-pharmacopoeial methods
  • All methods should be validated
  • Validation reports, including data conclusions
  • Stability of sample/standard solutions
  • Pharmacopoeial methods
  • Full or partial validation / verification of
    suitability
  • show validity for this formulation specificity
    important
  • Validation study - ICH guidelines
  • Q2A Q2B

45
3.9.3 Validation analytical methodsICH (Q2A)
table - validation parameters
Type of analytical procedure Type of analytical procedure Type of analytical procedure Type of analytical procedure
Characteristics ID impurities impurities Assay Incl. diss.
Characteristics ID quantitative limit Assay Incl. diss.
Accuracy
Precision
Repeatability
Iterm. precision
Specificity
Limit of detection ?
Limit of quantitation
Linearity
Range
46
3.10 Packaging 17/50
  • Container/closure system (immediate container)
  • Suitability for storage, transport, compatibility
  • protection against moisture, air, light if
    required
  • Detailed description, including liner/wadding
  • Specifications of parts in contact with product
  • Description
  • Identification (Typical IR - specific)
  • Drawings and critical dimensions
  • Outer packaging
  • Description, type of material

47
3.11 Stability testing 17/50 separate topic
  • One comment here
  • 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
  • from EMEA guidance CPMP/QWP/122/02

48
3.12 Container labelling 23/50
  • Outer packaging
  • where no outer packaging, on immediate packaging,
    e.g. HDPE bottle
  • Blisters and strips
  • All the elements as listed on page 23/50

49
3.13 SmPC 3.14 PIL 23/50
  • 3.13 Summary of product characteristics (SmPC)
  • To appear in WHOPAR
  • Changes to SmPC to be approved by WHO
  • See Annex 5 of guideline
  • 3.14 Patient information leaflet (PIL)
  • To appear in WHOPAR
  • In conformance with SmPC
  • See Annex 6 of guideline
  • From quality side, include (where appropriate)
  • Identification of dosage form in detail
  • Container description in detail
  • Storage requirements and approved shelf-life

50
Closing remarks
  • The joint efforts of manufacturers WHO
  • should cover all activities aimed at ensuring
    that the patient receives a product that meets
    established specifications and standards of
    quality, safety and efficacy
  • It concerns both
  • the quality of the products themselves and
  • anything that might affect quality,
  • including information supplied with the product
  • (derived from Health Action International
    (Africa) definition for QA)
  • Quality is built in by design, not tested in
Write a Comment
User Comments (0)
About PowerShow.com