session 3 validation, risk and criticality · session 3 validation, risk and criticality presented...

63
Session 3 Validation, Risk and Criticality Presented by Trevor Schoerie 14 May, 2014

Upload: others

Post on 09-Feb-2021

3 views

Category:

Documents


0 download

TRANSCRIPT

  • Session 3Validation, Risk and Criticality

    Presented by Trevor Schoerie

    14 May, 2014

  • Slide 2 © PharmOut 2014

    Validation, Risk and Criticality

    • In process validation we want to understand and control the variationnot the risk.

    • The “drug” benefit vs risk should have already been determined, (hopefully).

    “GAMP 5 – Risk Based ……”

    “We talk about QRM every time we meet!”

  • Slide 3 © PharmOut 2014

    “Process Variation”

    1. Sample Variation

    2. Analytical Variation?

    3. Process Variation – normally associate

    Is the sample representation of the batch?

    Blinded samples in lab = STD DEV

    Real process variation

  • Slide 4 © PharmOut 2014

    This session

    Using Risk to determine no of batches

    Risk vs criticality

  • Slide 5 © PharmOut 2014

    How we get to validation…..

    • New Chemical Entity *Stage 1 data

    • Generic manufacturer

    • Contract manufacturer

    • New manufacturing site – Tech. Transfer

    • Country where validation is a new requirement

    • Expansion of regulations, i.e. export to Australia

    No concurrent release of validation batches

    No retrospective validation

  • Slide 6 © PharmOut 2014

    Typical validation NCE – Stage 2a/b?

    Real life

    Start

    Registration / QbD

  • Slide 7 © PharmOut 2014

    How many PPQ batches?

    QTPP

    CQAs/

    CPPs

    Process Description

    Analytical Methods

    SOPs & Batch

    Records

    Design Reports

    FSE Qualified

    PVMP

    Training Completed

    Approved PV

    Protocols

  • Slide 8 © PharmOut 2014

    Knowledge vs # of PV batches

    Prior Knowledge Process Design

    Prior Knowledge

    PV

    Process Design PV

    Comprehensive Prior Knowledge may support fewer PV batches

    Limited Prior Knowledge may require more PV batches

  • Slide 9 © PharmOut 2014

    How many PPQ batches?

    This depends on the risk and the following elements:

    Based on science, experience and justified (documented).

    Based on statistics.

  • Slide 10 © PharmOut 2014

  • Slide 11 © PharmOut 2014

    How many PPQ batches?

    Step 2:Risk Assessment of Control

    Strategy

    Step 3:Determine Overall Residual

    Risk

    Step 4:Translate Overall Residual

    Risk into # of PPQ Batches

    Stage 1Assessment

    Stage 2 ActivitiesPrepare PPQ BatchesCompare results to Acceptance Criteria

    Risk Level AcceptableStep 1:Risk Assessment of Product

    Knowledge & Process Understanding

    Reference ISPE

  • Slide 12 © PharmOut 2014

    Risk Assessment of Stage 1

    Product Knowledge• Analytical understanding

    of product structure• Mechanistic

    understanding of product profile

    • Contextual understanding of CQA’s

    Process Understanding• Depth of understanding

    unit operations• Level of knowledge of

    process response to input variability

    • Process predictability• Understanding of scale

    Control Strategy• Application of

    process/product understanding to production controls

    • Control of raw material inputs

    • Equipment capability vsprocess requirements

    Assess each CQA

    Can use:• Risk Charts• Relative level of

    risk based on RPN

    • Other QRM tools

  • Slide 13 © PharmOut 2014

    Step 1: Risk Assessment of Product Knowledge

    Identification of CQA and impact of CQA variation on patient:

    • Level of understanding of product attributes (i.e. how a particular attribute affects patient safety & efficacy)

    Product Characterisation:

    • The strength of the link between the CQA and clinical performance

  • Slide 14 © PharmOut 2014

    Product Knowledge Risk Ranking

    Product Knowledge

    Factor

    Relative Risk RankingCharacteristics of ranking assignments

    Low Risk Medium Risk High Risk

    Identification of CQA and impact of CQA variation on patient

    • Physiochemical/ biological, pharmacokinetic knowledge, and QbDapproach used to design the formulation of drug product

    • Impact of variation on bioavailability explored & understood

    • CQAs identified and justified

    • Physiochemical/ biological, pharmacokinetic properties identified

    • Some exploration of impact of variation

    • Product specifications established from development trial and error

    • Impact of variation known only from evaluation of incidents

    Product Characterisation

    • Analytical method has direct measurable linkage to clinical performance

    • Complete product

    • Analytical method development based on mechanism of action for the therapeutic agent but linkage to clinical performance

    • Product characterisation measures quality against established empirical limits

  • Slide 15 © PharmOut 2014

    Process Understanding Risk Ranking

    Process Understanding

    Factor

    Relative Risk RankingCharacteristics of ranking assignments

    Low Risk Medium Risk High Risk

    Degree of process understanding/ unit operation

    -First principles understanding: based on an understanding of prevailing mechanisms and rationale

    -Casual knowledge: that based on what causes interrelationships between variables

    -Descriptiveknowledge: derived only from observation, reflecting basic facts

    Process predictably and modelling

    -Models based on first principles-Extension of empirical and mechanistic models-Highly predictable process and scale-up

    -Use of models derived from basic physical, chemical, biological or microbial mechanisms of observed phenomena-Sufficient knowledge to employ PAT methods, if applicable and desired

    -Primitive models reflecting only basic understanding of process and scale effects-Process predictability is questionable

    Table continued on the next slide

  • Slide 16 © PharmOut 2014

    Process Understanding Risk Ranking

    Process Understanding

    Factor

    Relative Risk RankingCharacteristics of ranking assignments

    Low Risk Medium Risk High Risk

    Process Response to input variability

    -Design space identified using multivariate data and statistical methods-Impact of material attributes on product quality explored extensively in development-Material specific CQAs identified and well understood or no material specific CQAs

    -Well-defined criticality for process based on multivariativeexperiments-Impact of material attributes on product quality explored to some degree-Material specific CQAs identified-full range of variability not explored in development

    -Partially defined, primarily through univariateexperimentation-Impact of materials attributes to product quality are minimally explored-Materials specific CQAs not identified

    Effects of scale -Highly predictable-dataacross different scales can be projected.

    -Predictable-data across scales can be projected

    -Unpredictable-Scale impact

  • Slide 17 © PharmOut 2014

    How many PPQ batches?

    Step 2:Risk Assessment of Control

    Strategy

    Step 3:Determine Overall Residual

    Risk

    Step 4:Translate Overall Residual

    Risk into # of PPQ Batches

    Stage 2 ActivitiesPrepare PPQ BatchesCompare results to Acceptance Criteria

    Risk Level AcceptableStep 1:Risk Assessment of Product

    Knowledge & Process Understanding

    Stage 1 ActivitiesAssessments of Product Knowledge and Process Understanding

  • Slide 18 © PharmOut 2014

    Step 2: Risk Assessment of Control Strategy

    • How the specifications were developed.Raw Materials Specifications

    • How easily the requirements for the process are accommodated by the manufacturing equipment.

    Equipment Capability vs. Process

    Requirements

    • How consistently the process has performed historically and during development studies.

    Experience with Process Performance

    • How the process is monitored and variability is detected

    Monitoring capability & detectability

  • Slide 19 © PharmOut 2014

    Control Strategy - Risk Ranking

    Control Strategy Factor

    Source of Potential

    Variability and/or Uncertainty

    Relative Risk RankingCharacteristics of ranking assignments

    Low Risk Medium Risk High Risk

    Raw Material Specifications

    -Different suppliers; different manufacturing processes, -Material attributes test method-Different batches-Basis for material specification-Specification wider than experience

    -Specifications of material attributes impacting product quality based on development data

    -Limited justification of specifications of material attributes

    -Specifications are not justified.-Compendial or supplier limits accepted without further investigation

  • Slide 20 © PharmOut 2014

    Control Strategy - Risk Ranking

    Control Strategy Factor

    Source of Potential

    Variability and/or Uncertainty

    Relative Risk RankingCharacteristics of ranking assignments

    Low Risk Medium Risk High Risk

    Equipment Capability vs.

    -Capability of equipment to

    -Comparison of parameter control

    -Comparison of control ranges from

    -Comparison of parameter control

    Process Requirements

    -Control operating parameters within acceptable ranges

    -Ranges from equipment qualification with process requirements indicates all parameters are well within equipment control capabilities and supported by qualification data

    -Equipment qualification with process requirements indicates marginal capability to meet requirements for a limited number of process parameters

    -Ranges from equipment qualification with process requirements indicates a significant number of parameters are similar to equipment control capabilities

  • Slide 21 © PharmOut 2014

    Control Strategy - Risk Ranking

    Control Strategy Factor

    Source of Potential Variability

    and/or Uncertainty

    Relative Risk RankingCharacteristics of ranking assignments

    Low Risk Medium Risk High Risk

    Experience with process performance to date

    -Variationobserved-Scaling effectsConsistency of past performance

    -Underlying cause(s) for variation is understood & addressed (or variation not observed during manufacture)-Impact of scale is well understood-Process has consistently performed as expected

    -Variation is managed empirically, but underlying causes are not well understood-Some understanding of scaling issues-Minor departures from expected results that were investigated and satisfactorily explained

    -Variation has been observed, but has not been successfully managed-Impact of scale changes has not been explored-Unexplainedfailure has been experienced

    Monitoring capability & detectability

    -Ability of monitoring tools & methods to detect variation

    -Attributes measured in real time at a sensitivity where performance variability is likely to be observed

    -Attributes measuredoff-line (after batch completion) at a sensitivity where performance is likely to be observed

    -Attribute measurement accuracy is inadequate

  • Slide 22 © PharmOut 2014

    Stage 2

    Step 2:Risk Assessment of Control

    Strategy

    Step 3:Determine Overall Residual

    Risk

    Step 4:Translate Overall Residual

    Risk into # of PPQ Batches

    Stage 1 ActivitiesAssessments of Product Knowledge and Process Understanding

    Stage 2 ActivitiesPrepare PPQ BatchesCompare results to Acceptance Criteria

    Risk Level AcceptableStep 1:Risk Assessment of Product

    Knowledge & Process Understanding

  • Slide 23 © PharmOut 2014

    Step 3: Determine overall residual risk

    The residual risk level reflects the confidence in performance of the commercial process:

    Residual Risk Level

    Description

    Severe (5)

    Multiple factors have high risk ratings

    High (4)

    Few factors have high risk ratings or all have medium risk rating

    Moderate (3)

    Medium risk level for multiple factors or high risk level for one factor

    Low (2)

    Medium risk level for a few factors, the others are low risk

    Minimal (1)

    Low risk level for all factors

  • Slide 24 © PharmOut 2014

    How many PPQ batches?

    Step 2:Risk Assessment of Control

    Strategy

    Step 3:Determine Overall Residual

    Risk

    Step 4:Translate Overall Residual

    Risk into # of PPQ Batches

    Stage 1Assess

    Stage 2 ActivitiesPrepare PPQ BatchesCompare results to Acceptance Criteria

    Risk Level AcceptableStep 1:Risk Assessment of Product

    Knowledge & Process Understanding

  • Slide 25 © PharmOut 2014

    Step 4: Translate overall residual risk into the number of PV Batches

    1. Based on rationales and experience

    2. Target Process Confidence and Target Process Capability

    • Statistical-includes measures of variability & confidence level

    3. Expected Coverage

    • Statistical-includes measure of probability of batch success rate

    Note: Other approaches may be appropriate

  • Slide 26 © PharmOut 2014

    How many PV batches?

    Approach 1:Rationales and

    experience

    Approach 2:Target process confidence &

    target process capability

    Step 4:Translate Overall

    Residual Risk into # of

    PPQ Batches

    2 Approaches to Translate Overall Residual Risk into # of PV Batches

  • Slide 27 © PharmOut 2014

    Approach 1: Rationales and Experience

    ResidualRisk Level

    Number of Batches

    Rationale

    Severe (5)

    Not Ready for PV

    Encourage additional development to reduce risk level

    High (4)

    10 High # of consecutive successful batches unlikely if controls are not adequate

    Moderate (3)

    5 Increased residual risk addressed by preparing 2 additional PV batches to provide further demonstration of process consistency

    Low (2)

    3 Knowledge & Control Strategy regarded as sufficient. 3 PPQ batches has historically been appropriate for demonstrating process consistency for many low-risk processes

    Minimal (1)

    1-2 Minimal residual risk with less than 3 PPQ batches required, e.g. for verifying specific controls associated with a well-understood change to a process

  • Slide 28 © PharmOut 2014

    Approach 2: Target process confidence & target process capability

    • Process Capability (CpK) is used as a measure of the capability of the process to consistently meet the quality requirements

    • Assumption: CpK ≥1 as a starting point for assessing the capability of a process undergoing validation

  • Slide 29 © PharmOut 2014

    Approach 2: Target process confidence & target process capability

    • Used as a measure for level of confidence needed in the CpK and thereby as a degree of assurance

    • High level of confidence in the CpK can be built only with time and experience (during Stage 3)

    • Residual Risk Level used to define the confidence needed at completion of Stage 2.

  • Slide 30 © PharmOut 2014

    Approach 2: Target process confidence & target process capability

    ResidualRisk Level

    TargetConfidence

    Comments

    Severe (5) N/A Major gaps in knowledge & understanding.Additional effort on product/process/control strategy development may be necessary. High confidence level needed to provide high degree of assurance.

    High (4) 97%

    Moderate (3)

    95% Target confidence levels designed to provide reasonable assurance of process capability. Higher confidence levels would be achieved during Stage 3.Low (2) 90%

    Minimal (1) N/A High “confidence” based on existing understanding and capability of control strategy. Does not require additional assurance during PV beyond demonstration that commercial systems and procedures are appropriate.

  • Slide 31 © PharmOut 2014

    Approach 2: Target process confidence & target process capability

    Residual Risk Level

    Min # of batches

    Target Confidence for CpK 1.0

    Acceptance Criteria

    Readily Pass Calculated

    CpK

    Marginally Pass Calculated CpK

    Fail Calculated CpK

    Severe (5) Not Ready for PPQ

    N/A

    High (4) 11 97%

    ≥ 1.6≥ 1.0and

    < 1.6< 1.0

    Moderate (3)

    8 95%

    Low (2) 5 90%

    Minimal (1) 1-3 N/A

    • Based on a Target CpK of 1.0 and an actual CpK of 1.6. • May include clinical manufacture, demonstration, or other at-scale lots.

    Assumes process will be under statistical control & data will show normal distribution.

  • Slide 32 © PharmOut 2014

    Approach 2: Target process confidence & target process capability

    PPQ Outcome Pass / Fail Other Considerations

    Impact on Initial CPV Sampling Approach*

    Readily Pass calculated CpK

    Pass N/A Supports Stage 3 routine sampling

    Marginally Pass calculated CpK

    Pass All input/output parameters within range

    Consider enhanced monitoring for CQA’s not meeting “Readily Pass”

    Fail calculated CpK

    Investigate -Parameter values-Intra-lot CpK-Probability of detection-Process improvement options

    Consider enhanced monitoring; May include some testing beyond PPQ.

    *PPQ outcome is one consideration in establishing CPV Plan

  • Slide 33 © PharmOut 2014

    This session

    Using Risk to determine no of batches

    Risk vs criticality

  • Slide 34 © PharmOut 2014

    FMEA - recap

    • Study the Failure Mode and Effect Analysis

    • It involves reviewing as many components, assemblies, and subsystems as possible to identify failure modes, and their causes and effects.

  • Slide 35 © PharmOut 2014

    Ishikawa – Cause and Effect Diagram

  • Slide 36 © PharmOut 2014

    Quality Risk Management (QRM)

    • This session will take a step back and look at the basics

    • We will ponder some topics:

    • Is the “Criticality Assessment” a “Variability Assessment” when discussing CQAs/CPPs for Qualification & Validation activities?

    • Do we throw out our innate understanding of the hazard because we are “FMEA” driven?

    FMEA does not help identify sources of variation

  • Slide 37 © PharmOut 2014

    ICH Q9 Quality Risk Management (& Annex 20 of PIC/S PE 009-8)

    • ICH Q9 explains the “What” of QRM [10 pages]

    • Annex I of ICH Q9 provides concepts and ideas on the “How to”-formal and informal [4 pages]

    • Annex II of ICH Q9 details the potential “Where” of QRM [5 pages]

    • A “roadmap” is presented to us within the document, but do we follow it for Qualification & Validation activities?

    • For example, do we make use of QRM tools other than FMEA (or FMECA) effectively?

  • Slide 38 © PharmOut 2014

    Quality Risk Management (QRM)

    • Risk is defined as the combination of the probability of occurrenceof harm and the severityof that harm

    • Harm: Damage to health, including the damage that can occur from loss of product quality or availability.

    • Severity: A measure of the possible consequences of a hazard.

    Begin the QRM Process

    Risk Identification

    Risk Analysis

    Risk Evaluation

    Risk Reduction

    Risk Acceptance

    Risk Assessment

    Risk Control

    Output of the QRM Process

    Review Events

    Risk Review

    QRMTools

    Risk C

    om

    mu

    nicatio

    n

  • Slide 39 © PharmOut 2014

    Quality Risk Management (QRM)

    Wet Floor (Hazard: The potential source of harm)

    Broken Leg(Harm: Damage to health, including the damage that can occur from loss of product quality or availability)

    Severity: A measure of the possible consequences of a hazard.

    Risk is defined as the combination of the probability of occurrence of harmand the severity of that harm

  • Slide 41 © PharmOut 2014

    Deductive vs Inductive Reasoning

    Destroyed my car.How did this happen?

    Intoxicated?What will happen if I

    drive?

    Inductive (forward logic)Deductive

    ?

    FMEAPHA

    FTA STA C&E Analysis

  • Slide 42 © PharmOut 2014

    Inductive Reasoning

    Outcome

    Hypothesis

    Observation

    Confirmation

    Risk Identification

    Risk Analysis

    Risk Evaluation

    Risk Reduction

    Risk Acceptance

    “Top-down" “Think up” Failures!

    More Proactive?

  • Slide 43 © PharmOut 2014

    Deductive Reasoning

    Theory

    Tentative Hypothesis

    Pattern

    ObservationRisk

    Identification

    Risk Analysis

    Risk Evaluation

    Risk Reduction

    Risk Acceptance

    “Bottom-up" “Narrow down”

    Failure!More Reactive?

  • Slide 44 © PharmOut 2014

    Inductive vs Deductive QRM

    QRM Tool Inductive Deductive “Complexity”

    FMEA Yes No Medium

    FMECA Yes No Medium

    FTA No Yes High

    HACCP Yes Yes Low

    HAZOP Yes No Medium

    PHA Yes No Low

    Risk Ranking and Filtering

    Yes Yes Low

    Supporting Statistical Tools

    Yes Yes Low

  • Slide 45 © PharmOut 2014

    Appendix I: Risk Management Methods & Tools

    • General overview-references some primary tools

    “It is important to note that no one tool or set of tools is applicable to every situation in which a QRM procedure

    is used”

    FMEA FMECA FTA HACCP

    HAZOP PHARisk Ranking & Filtering

    Supporting Stats Tools

  • Slide 46 © PharmOut 2014

    Appendix I: Risk Management Methods & ToolsQRM Tools Input Process Output

    FMEA Relies on product & process understanding. Manageable process steps.

    Once potential failure modes are established, risk reduction can be used to eliminate, contain, reduce or control the potential failures.

    Summarises modes of failure, factors causing these failures and the likely effects of these failures.

    FMECA Relies on product & process understanding. Manageable process steps.

    Once potential failure modes are established, risk reduction can be used to eliminate, contain, reduce or control the potential failures.

    The output of an FMECA is a relative risk “score” for each failure mode, which is used to rank the modes on a relative risk basis.

    FTA System (or sub-system) failures

    Evaluates system (or sub-system) failures one at a time but can combine multiple causes of failure by identifying causal chains.

    The output of an FTA includes a visual representation of failure modes.

  • Slide 47 © PharmOut 2014

    Failure Mode Effects Analysis (FMEA)

    Item or process

    Step

    Potential Failure Mode

    Potential Effect(s) of Failure

    Se

    ve

    rity

    Potential Cause(s)

    Occu

    rren

    ce

    Current Controls

    De

    tectio

    n

    RP

    N

    Recommended Action

    Responsibility & Target Date

    Action Taken

    Se

    ve

    rity

    Occu

    rren

    ce

    De

    tectio

    n

    RP

    N

    FMECA: Extended to incorporate an investigation of the degree of severity of the consequences, their respective probabilities of occurrence and their detectability

  • Slide 48 © PharmOut 2014

    Appendix I: Risk Management Methods & ToolsQRM Tools Input Process Output

    HACCP Product and process understanding.

    It is a structured approach that applies technical & scientific principles to analyze, evaluate, prevent, and control the risk or adverse consequence(s) of hazard(s)

    Risk management information that facilitates monitoring of critical points not only in the manufacturing process but also in other life cycle phases.

    HAZOP All processes & safety hazards.

    It is a systematic brainstorming technique for identifying hazards using so-called “guide-words”.

    As is the case with HACCP, the output of a HAZOP analysis is a list of critical operations for risk management.

    PHA Product, process and facility design information

    1) Identification, 2) Evaluation 3) Ranking, and 4) Remediation

    Typically, hazards identified in the PHA are further assessed with other risk management tools such as those in this section.

  • Slide 49 © PharmOut 2014

    Preliminary Hazard Analysis (PHA)

    • Early in the development: little information on design details or operating procedures will often be a precursor to further studies

    • For product, process and facility design

    • Further assessed with other risk management toolsHazards Arising from Product Design

    Hazard Investigation/ Controls

    Severity (S)

    Frequency (F)

    Impact (SxF)

  • Slide 50 © PharmOut 2014

    Appendix I: Risk Management Methods & ToolsQRM Tools Input Process Output

    Risk Ranking and Filtering

    Risk ranking and filtering can be used to prioritize manufacturing sites for inspection/audit by regulators or industry.

    Forms a single relative risk score that can then be used for ranking risks. “Filters,” in the form of weighting factors or cut-offs for risk scores, can be used to scale or fit the risk ranking to management or policy objectives.

    Risk ranking methods are particularly helpful in situations in which the portfolio of risks and the underlying consequences to be managed are diverse and difficult to compare using a single tool.

    Supporting Statistical Tools

    Statistical data They can enable effective data assessment, aid in determining the significance of the data set(s), and facilitate more reliable decision making.

    Control Charts, Design of Experiments (DOE), Histograms, Pareto Charts, Process Capability Analysis

  • Slide 51 © PharmOut 2014

    Appendix II: Potential Applications for QRM

    In II.1: Integrated Quality Management

    • To interpret monitoring data (e.g., to support an assessment of the appropriateness of revalidation or changes in sampling).

    • To determine appropriate actions preceding the implementation of a change, e.g., additional testing, (re)qualification, (re)validation or communication with regulators.

  • Slide 52 © PharmOut 2014

    Appendix II: Potential Applications for QRM

    In II.4: Facilities, Systems & Equipment

    • To determine the scope and extent of qualification of facilities, buildings, and production equipment and/or laboratory instruments (including proper calibration methods).

    • To determine acceptable (specified) cleaning validation limits.

  • Slide 53 © PharmOut 2014

    Appendix II: Potential Applications for QRM

    In II.4: Facilities, Systems & Equipment

    To select the design of computer hardware and software (e.g., modular, structured, fault tolerance);

    To determine the extent of validation, e.g.,

    • identification of critical performance parameters; selection of the requirements and design;

    • code review;

    • the extent of testing and test methods;

    • reliability of electronic records and signatures.

  • Slide 54 © PharmOut 2014

    Appendix II: Potential Applications for QRM

    II.6 Production

    • To identify the scope and extent of verification, qualification and validation activities (e.g., analytical methods, processes, equipment and cleaning methods;

    • To determine the extent for follow-up activities (e.g., sampling, monitoring and re-validation);

    • To distinguish between critical and non-criticalprocess steps to facilitate design of a validationstudy.

  • Slide 55 © PharmOut 2014

    Uses in Qualification & Validation

    QRM ToolsTools

    Uses in Q&V

    FMEA All Q&V Stages-prioritize potential risks and monitor the effectiveness of risk control activities.

    FMECA All Stages-Prioritize potential risks and monitor the effectiveness of risk control activities. Understand the impact of detectability

    FTA Establish the pathway to the root cause of the failure during Q&V. Is useful both for risk assessment and in developing monitoring programs as an output of Q&V.

    HACCP Used to identify and manage risks associated with physical, chemical and biological hazards (including microbiological contamination).

    HAZOP This facilitates regular monitoring of critical points in the manufacturing process. Used early in the design stage. Q&V input.

    PHA Used early in the development of a project when there is little information on design details or operating procedures; thus, it will often be a precursor to further studies. Useful “first cut” for Q&V.

    Risk Ranking and Filtering

    Risk ranking is useful when management needs to evaluate both quantitatively-assessed and qualitatively-assessed risks within the same organizational framework.

    Stats Tools They can enable effective data assessment throughout Q&V activities

  • Slide 56 © PharmOut 2014

    QRM vs Impact Assessment Example

    “Top Down”

    Critical Quality Attributes

    Critical Process Parameters

    System

    Subsystem

    Component

    End

    End

    Product carryover exceeds acceptance criteria

    Cleaning Agent Concentration

    CIP System

    Chemical Addition

    Chemical Feed Pump

    “Bottom Up”

    From ISPE GPG Applied Risk Management for C&Q

  • Slide 57 © PharmOut 2014

    QRM vs Impact Assessment

    Characteristics Impact Assessment (Baseline® Guide 5)

    Formal Risk Assessment

    “Top Down” or “Bottom Up” “Bottom Up” “Top Down”

    Ability to identify specific process risks Low High

    Ability to identify specific Critical Aspects

    Low High

    Ability to prioritise Qualification efforts Medium High

    Cost in time and resources Medium High

    Need for SMEs experienced in the tool to be used

    Low High

    Acceptability to regulators Medium High

    Usefulness as “lifecycle” document in future

    Low High

    From ISPE GPG Applied Risk Management for C&Q

  • Slide 58 © PharmOut 2014

    “Criticality” in Annex 15

    • “It is a requirement of GMP that manufacturers identify what validation work is needed to prove control of the critical aspects of their particular operations.”

    • “The protocol should specify critical steps and acceptance criteria.”

    • “Evidence should be available to support and verify the operating parameters and limits for the critical variables of the operating equipment.”

    • “Risk analysis: Method to assess and characterise the critical parameters in the functionality of an equipment or process.”

  • Slide 59 © PharmOut 2014

    Going from QTPP to CQA

    • We assume that during product development the clinicians and toxicologists have established that these parameters are high risk to the patient.

    • So we are looking at reducing the variability of these attributes.

  • Slide 60 © PharmOut 2014

    Connecting a CQA to a CPP

    • The manufacturer during product development and based on prior product and process knowledge establishes that these process parameters have a high impact on variability.

    • “Validation” looks at ways to provide confidence that the variability is controlled.

  • Slide 65 © PharmOut 2014

    Ishikawa – Cause and Effect Diagram

  • Slide 66 © PharmOut 2014

    The PharmOut “STIC-man”!

    Summary Table from Ishikawa Criticality

  • Slide 67 © PharmOut 2014

    Activity 3: QRM

    Please complete the Blue Sheets as a group and return them to us

    Please assign a Speaker/Scribe at

    each table

    If there are any questions, please ask!

  • Slide 68 © PharmOut 2014

    Thank you for your time.Questions?

    Trevor Schoerie

    [email protected]

    Lead Consultant

    www.pharmout.net