Workshop on Quality Assurance and GMP of Multisource HIV/AIDS medicines - PowerPoint PPT Presentation

1 / 60
About This Presentation
Title:

Workshop on Quality Assurance and GMP of Multisource HIV/AIDS medicines

Description:

Workshop on Quality Assurance and GMP of Multisource HIV/AIDS medicines QUALIFICATION and VALIDATION II. J nos Pog ny, pharmacist, PhD, consultant to WHO – PowerPoint PPT presentation

Number of Views:233
Avg rating:3.0/5.0
Slides: 61
Provided by: whoIntpre
Category:

less

Transcript and Presenter's Notes

Title: Workshop on Quality Assurance and GMP of Multisource HIV/AIDS medicines


1
Workshop on Quality Assurance and GMP of
Multisource HIV/AIDS medicines
QUALIFICATION and VALIDATION II.
  • János Pogány, pharmacist, PhD,
  • consultant to WHO
  • Shanghai, 01 March 2005
  • E-mail pogany_at_axelero.hu

2
GMP - 4.11 Analytical methods, computers and
cleaning procedures
  • It is of critical importance that particular
    attention is paid to the validation of analytical
    test methods, automated systems and cleaning
    procedures.
  • Validation of analytical procedures used in the
    examination of pharmaceutical materials (WHO
    Expert Committee on Specifications for
    Pharmaceutical Preparations. 32nd Report. Geneva,
    WHO, 1992 (WHO Technical Report Series, No. 823).
  • Text on Validation of Analytical Procedures Q2A
    (1994)
  • Validation of Analytical Procedures Methodology
    Q2B (1996)
  • ICH Harmonized Tripartite Guidelines

3
Characteristics of analytical procedures (1)
  • Accuracy (also termed trueness)
  • Precision
  • Repeatability
  • intermediate precision (within-laboratory
    variation)
  • reproducibility (inter-laboratory variation)
  • Robustness, ruggedness

4
Characteristics of analytical procedures (2)
  • Linearity
  • Range
  • Specificity (selectivity)
  • Sensitivity (versus robustness)
  • Limit of detection
  • Limit of quantitation

5
Accuracy and precision
Inaccurate and imprecise
Accurate
Accurate and precise
Precise
6
Classes of analytical tests
  • The objective of validation of an analytical
    procedure is to demonstrate that it is suitable
    for its intented purpose.
  • Class A To establish identity
  • Class B To detect (Bd) and quantitate (Bq)
    impurities
  • Class C To determine quantitatively the
    concentration, or assay
  • Class D To assess characteristics
  • Other classes not covered in the guides

7
Criteria for analytical classes
Criteria A Bq Bd C D
Accuracy X X X
Precision X X X
Robustness X X X X X
Linearity and range X X X
Specificity X X X X X
Limit of detection X
Limit of quantitation X
8
General requirements
  • Qualified and calibrated instruments
  • Documented methods
  • Reliable reference standards
  • Qualified analysts
  • Sample integrity
  • Change control (e.g., synthesis, FPP composition)

9
HPLC Method Development and Validation for
Pharmaceutical Analysis by Ghulam A. Shabir
  • Pharmaceutical Technology Europe, 1 March 2004

10
Prequalification requirements
  • Analytical method validation is required by WHO
    for the prequalification of product dossiers.
    Non-compendial ARV APIs and FPPs were/are tested
    with methods developed by the manufacturer.
  • Analytical methods should be used within GMP and
    GLP environments, and must be developed using the
    protocols and acceptance criteria set out in the
    ICH guidelines Q2A and Q2B.

11
HPLC system
12
Linearity and range
13
ICH requirements
  • Concentration range 0.0250.15 mg/mL (25150 of
    the theoretical concentration in the test
    preparation, n3)
  • Regression equation was found by plotting the
    peak area (y) versus the analyte concentration
    (x) expressed in mg/mL y 3007.2x 4250.1 (r2
    1.000).
  • The regression coefficient demonstrates the
    excellent relationship between peak area and
    concentration of analyte.
  • The analyte response is linear across 80-120 of
    the target progesterone concentration.

14
Accuracy
The data show that the recovery of analyte in
spiked samples met the evaluation criterion for
accuracy (100 2.0 across 80120 of target
concentrations).
15
Specificity
16
Specificity
  • An example of specificity criterion for an assay
    method is that the analyte peak will have
    baseline chromatographic resolution of at least
    2.0 from all other sample components.
  • In this study, a weight of sample placebo
    equivalent to the amount present in a sample
    solution preparation was injected to demonstrate
    the absence of interference with progesterone
    elution. Former slide demonstrates specificity.

17
Repeatability
  • The repeatability precision obtained by one
    analyst in one laboratory was 1.25 RSD for the
    analyte and, therefore, meets the evaluation
    criterion of RSD 2.

18
Intermediate precision
19
Limit of detection
  • The limit of detection (LOD) is defined as the
    lowest concentration of an analyte in a sample
    that can be detected, not quantified. It is
    expressed as a concentration at a specified
    signalnoise ratio (SNR), usually between 3 and
    21.
  • In this study, the LOD was determined to be
    10ng/mL with a signalnoise ratio of 2.91.

20
Limit of quantitation
  • The limit of quantitation (LOQ) is defined as the
    lowest concentration of an analyte in a sample
    that can be determined with acceptable precision
    and accuracy under the stated operational
    conditions of the method. The ICH has recommended
    a signal noise ratio (SNR) of 101.
  • The LOQ was 20 ng/mL with a signalnoise ratio of
    10.2. The RSD for six injections of the LOQ
    solution was 2.

21
LOD, LOQ and SNT
  • Limit of Quantitation (LOQ)
  • Limit of Detection (LOD)
  • Signal to Noise Ratio (SNR)

Peak BLOQ
Peak ALOD
noise
Baseline
22
Analytical solution stability
Standard and sample solutions stored in a capped
volumetric flask on a lab bench under normal
lighting conditions for 24 h were shown to be
stable with no significant change in progesterone
concentration during this period.
23
Conclusion of validation study
  • The relation between concentration and peak area
    is linear in the range from 25 to 150. R1.
  • Accuracy shows a mean with a RSD of 0.59 over the
    the range from 50 to 150.
  • The method is specific in the given formulation.
  • Repeatability and intermediate precision are well
    within the 2 RSD criterion.
  • LOD is 10 ng/mL and LOQ is 20 ng/mL.
  • The analytical solution is stable for 24 hours.

24
Workshop on Quality Assurance and GMP of
Multisource HIV/AIDS medicines
CLEANING VALIDATION
  • János Pogány, pharmacist, Ph.D.
  • Shanghai, 03 March 2005
  • E-mail pogany_at_axelero.hu

25
Subjects for Discussion
  • Regulatory background
  • General considerations
  • Cleaning validation guideline Canada
  • An illustrative approach to the cleaning
    validation of antiretroviral (ARV) active
    pharmaceutical ingredient(s) API(s
  • A case study from literature
  • Conclusions

26
WHO GMP
  • 4.11 It is of critical importance that particular
    attention is paid to the validation of ...
    cleaning procedures.
  • 16.11 Contamination of ... a product by another
    material or product must be avoided. This risk of
    accidental cross contamination arises from ...
    products in process, from residues on equipment.
    Among the most hazardous contaminants are highly
    sensitizing materials ... and highly active
    materials.

27
WHO GMP
  • 16.15 Before any processing operation is started,
    steps should be taken to ensure that the work
    area and equipment are clean.
  • 16.18 Time limits for storage of equipment after
    cleaning and before use should be stated and
    based on data.

28
Why do we validate cleaning processes?
  • The cleaning process is an integral part of the
    pharmaceutical manufacturing process.
  • Industry should view cleaning of equipment as the
    first manufacturing step. (It will have effect on
    the safety, efficacy and quality of the batch to
    be manufactured.)
  • A cleaning process must be chosen based on
    products (e.g., ARVs, solid dosage forms),
    objectives, resources, and limitations within
    each manufacturing company.

29
Pharmaceutical Process Validation Second
Edition, Revised and Expanded, edited by Ira R.
Berry and Robert A. Nash, Marcel Dekker, Inc.,
New York Basel Hong Kong (1993).
  • GENERAL
  • CONSIDERATIONS

30
Potential Contaminants
  • Chemical contamination
  • Product residues
  • Decomposition residues
  • Cleaning or disinfecting agent residues
  • Microbiological contamination
  • Bacteria, moulds, pyrogens
  • Unintended materials
  • Airborne (particulate) matter
  • Lubricants, ancillary material (e.g. pieces of
    brushes)

31
Manual Cleaning Procedures
  • Equipment disassembly (if required)
  • Prewash and inspection (most visible material
    removed)
  • Wash (cleaning agent, temperature, multiple steps
    until visually clean)
  • Initial rinses (rinse water, temperature)
  • Final rinse (minimum dissolved solids,
    microorganisms)
  • Reassembly (if required)

32
Automated Cleaning Procedures
  • Clean-in-place (CIP) systems (dishwasher-type
    equipment)
  • portable (tank and pump assemblies on wheels)
  • stationery, cabinet-type
  • Control system qualification (reproducibility,
    water temperature control)
  • Sampling (sampling port, pause capability)
  • Material supply (hard-plumbed supply lines,
    volume and dispensing controls, potential impact
    of long storage periods)

33
Documentation and Traceability
  • Equipment identification
  • Equipment use, maintenance and cleaning records
  • Labeling
  • Cleaning equipment maintenance and calibration
  • Utilities (water for injection (WFI), purified
    water, steam and compressed air systems)
    qualified and validated.
  • Standard Operating Procedure(s) SOP(s)
  • Personnel training

34
Cleaning Materials and Tools
  • Solvents (source and quality controlled)
  • Cleaning agents (acids, bases, surfactants, etc.,
    qualified type and brand QC controlled)
  • Ancillary utilities (steam and compressed air
    qualified)
  • Scrubbing agents (compression of placebo tablets
    to clean punches and dies)
  • Cleaning tools (standard sets of brushes, rags,
    sponges)
  • Equipment (thermometers, CIP systems consisting
    of tanks, metering pumps, heat exchangers, etc.
    maintaned and kept in calibrated status)

35
Frequency of Cleaning
  • Cleaning between batches of the same product
    (abbreviated procedures)
  • Cleaning between batches of different products
  • Cleaning after maintenance
  • Cleaning after accidental contamination

36
Cleaning Validation Guidelines,Health Products
and Food Branch Inspectorate, Canada
http//www.hc-sc.gc.ca/hpfb-dgpsa/inspectorate/cl
ean_val_gui_entire_e.html
  • PRESENTATION IS LIMITED TO SOLID PHARMACEUTICAL
    DOSAGE FORMS

37
Validation of cleaning processes
  • Equipment cleaning validation may be performed
    concurrently with actual production steps during
    process development and clinical manufacturing.
    Validation programs should be continued through
    full-scale commercial production.
  • All pertinent parameters should be checked to
    ensure the process, as it will ultimately be run
    is validated. Therefore, if critical temperatures
    are needed to effect cleaning, then these should
    be verified. Any chemical agents added should be
    verified for type as well as quantity. Volumes of
    wash and rinse fluids, and velocity measurements
    for cleaning fluids should be measured as
    appropriate.

38
Validation of cleaning processes
  • Validation of cleaning processes should be based
    on a worst-case scenario including
  • challenge of the cleaning process to show that
    the challenge soil can be recovered in sufficient
    quantity or demonstrate log removal to ensure
    that the cleaning process is indeed removing the
    soil to the required level, and
  • the use of stress cleaning parameters such as
    overloading of contaminants, overdrying of
    equipment surfaces, minimal concentration of
    cleaning agents and/or minimum contact time of
    detergents.

39
Validation of cleaning processes
  • At least three (3) consecutive applications of
    the cleaning procedure should be performed and
    shown to be successful in order to prove that the
    method is validated.

40
Approach for setting limits
  1. Product specific cleaning validation for all
    products
  2. Grouping into product families and choosing a
    worst case product
  3. Grouping into risk categories (e.g., very soluble
    products, similar potency, highly toxic products
    or difficult to detect)
  4. Setting limits on not allowing more than a
    certain fraction of carryover
  5. Different safety factors for different dosage
    forms.

41
Carry-over of product residues
  • NMT 0.1 of the normal therapeutic dose of any
    product to appear in the maximum daily dose of
    the following product (may not be acceptable for
    parenterals).
  • NMT 10 ppm of any product to appear in another
    product (may not be acceptable for parenterals).
  • No quantity of residue to be visible on the
    equipment after cleaning procedures are
    performed. (Spiking studies should determine the
    concentration at which most active ingredients
    are visible.)

42
Carry-over of product residues
  • Residues levels that do not interfere with
    subsequent manufacturing processes.
  • For certain allergenic ingredients, penicillins,
    cephalosporins or potent steroids and cytotoxics,
    the limits should be below the limit of detection
    by best available analytical methods. In practice
    this may mean that dedicated plants are used for
    these products.
  • Acceptable limits should be defined for detergent
    residues after cleaning (there is no normal
    therapeutic dose, thus e.g. the limit of
    detection of the most toxic component).

43
Analytical methods
  • The analytical methods used to detect residuals
    or contaminants should be specific and be
    validated before the cleaning validation study is
    carried out.
  • The specificity and sensitivity of the analytical
    methods should be determined.
  • The analytical method and the percent recovery of
    contaminants should be challenged in combination
    with the sampling method(s).

44
Sampling and related issues
  • Direct surface sampling (swab method)
  • Indirect sampling (use of rinse solutions)
  • Indirect testing such as monitoring conductivity
    may be of some value
  • In terms of cross-contamination, the main concern
    is residue left on the internal product-contact
    surfaces of the manufacturing equipment.

45
An Illustrative Approach to Cleaning Validation
  • ANTIRETROVIRAL
  • FPP(s)

46
Cleaning validation (master) plan
  • Validation plan is based on risk analysis.
  • Cleaning of individual pieces of the
    manufacturing and packaging equipment is
    validated with products selected as the worst
    case.
  • The three regulatory consecutive batches can be
    extended to include potentially the last batches
    of one or more campaign productions
  • Water solubility, toxicity and risk analysis data
    of all ARV APIs.

47
Risk Analysis
RISK FACTORS
48
Illustrative Indicators for Toxicity
Composite toxicity indicators may take into
account high activity, hypersensitizing
indicators, etc.
49
Illustrative Categories for Solubility
50
Illustrative Risk Analysis of ARV APIs
51
Pharmaceutical Technology Europe, 1 February
2004Griet Van Vaerenbergh
  • Cleaning Validation Practices Using a One-Pot
    Processor

52
Summary
  • This article describes the use of a one-pot
    processor for the cleaning and cleaning
    validation of two drug compounds water-soluble
    theophylline and water-insoluble mebendazole.
    Both substances were produced using wet
    granulation and microwave drying, after which the
    processor was cleaned using its clean-in-place
    (CIP) system. Swab samples were taken from areas
    considered critical during processing and
    analysed for remains of active ingredient. It was
    concluded from the results that the processor's
    CIP system is capable of removing both APIs to a
    level well within accepted regulations.

53
One-pot processor
54
Acceptance criteria
  • 10-ppm criterion
  • absolute mass criterion NMT 1 µg/cm2
  • for residual detergent traces the conductivity
    of the final rinsing water should be lower than
    the conductivity of a 11000 dilution of the
    detergent solution.

55
Acceptable quantity of an API per swab
  • Initial trials on the swab determined that the
    theophylline recovery was between 95100.
    Nevertheless, the Factor 2 for swab yield was
    maintained in the formula for calculating the
    acceptance criteria, to account for any operator
    influence.

56
Theophylline sample analysis
57
Mebendazole sample analysis
58
Study conclusions
  • This study has shown that the CIP system of this
    one-pot processor is capable of removing both
    water-insoluble mebendazole and water-soluble
    theophylline from the system to a level
    significantly less than acceptable maxima.
    Although certain areas show a larger variation in
    results than others, the reproducibility of the
    cleaning cycle can be considered good, as the
    results for all areas were always consistent.

59
Main Points Again
  • Validation of equipment cleaning processes is
    critical to safety, efficacy and quality of FPPs.
  • There is no generally accepted approach to
    cleaning validation.
  • One possible approach is risk analysis and
    selection of worst case for each item of
    equipment.
  • CIP equipment must be qualified and the cleaning
    processes must be validated.

60
THANK YOU
??!
Write a Comment
User Comments (0)
About PowerShow.com