ayush reportnew 1 (2)

Embed Size (px)

Citation preview

  • 8/6/2019 ayush reportnew 1 (2)

    1/41

    (1)

    Pranveer singh institute of technology, kanpur

    Institute of pharmacy

    CERTIFICATE

    This is to certify that the project work entitled is a project work done

    by Mahendra Kr. Verma under the supervision ofMr. Swatantra

    Kushwaha, Assit. Professor, institute of pharmacy,PSIT,Kanpur.

    Date: Mr. A. K. RAI

    DIRICTOR OF PHARMACY

    Place : PSIT, Kanpur

  • 8/6/2019 ayush reportnew 1 (2)

    2/41

    (2)

    LIST OF CONTENTS

    Serial

    no.

    Content Page no.

    1 Introduction 1

  • 8/6/2019 ayush reportnew 1 (2)

    3/41

    (3)

    Introduction

    A process is a series of interrelated functions and activities using a

    variety of specified actions and equipment which is designed to produce

    a defined result. To validate the reproducibility and consistency of a

    process, the full defined process is carried out using validated

    equipment, under the established procedure usually at least 3 times The

    process must successfully and consistently meet all acceptance criteria

    each time to be considered a validated process. In many cases, "worst

    case" conditions are used for the validation to ensure that the process is

    acceptable in the extreme

    case. Sometimes worst case conditions for systems can only really be

    tested over time and hence must be evaluated using a rigorous long

    term monitoring programme.

    Examples of processes which must be validated in pharmaceutical

    manufacturing are:

    Cleaning,Sanitization,Fumigation,Depyrogenation,Sterilization

    Sterile filling,Fermentation,Bulk production,Purification

    Filling, capping, sealing Lyophilization

    This guideline outlines the principles and approaches that SFDA

    considers important for the process validation of manufacturing

    processes. It provides that activities align with process validation during

    the product and process development, including all terms/ definitions

    used in the validation.It should be noted that the recommendations

    suggested in this guideline are not intended as requirements under all

    circumstances as the requirement of

  • 8/6/2019 ayush reportnew 1 (2)

    4/41

    (4)

    the SFDA. Alternate means, scientifically justified and documented are

    equally acceptable.Each process to be validated must be a specific

    process clearly described in a Master Formula or in an SOP. It is very

    important that the specifications for a process undergoing validation be

    pre-determined. It is also important that for all critical processingparameters for which specifications have been set, there must be

    equipment to measure all of the separateness during the validation

    study.

    Process Validation studies examine a process under normal operating

    conditions to prove that the process is in control. Once the process has

    been validated, it is expected that it remains in control, provided no

    changes are made.

    In the event that modifications to the process are made, or problemsoccur, or equipment or systems involved in the process are changed, a

    re-validation of the process would be required. Very often validation

    studies require that more measurements are made than are required for

    the routine process. The validation must prove the consistency of the

    process and therefore must assess the efficiency and effectiveness of

    each step to produce its intended outcome.The controls and tests and

    their specifications must be defined. To be considered validated, the

    process must consistently meet all specifications at all steps throughout

    the procedure at least three times

    Objective and purpose

    Process validation of pharmaceutical manufacturing process especially

    tablet manufacturing process with special reference to the

    requirements stipulated by the US Food and Drug Administration (FDA).

    Quality is always an imperative prerequisite when we consider any

    product. Therefore drugs must be manufactured to the highest quality

    levels. End product testing by itself does not guarantee the quality of theproduct. Quality assurance techniques must be used to build the quality

    in to the product at every step and not just tested for at the end. In

    pharmaceutical industry, Process validation performs this task to build

    the quality into the product because according to ISO

  • 8/6/2019 ayush reportnew 1 (2)

    5/41

    (5)

    9000:2000, it had proven to be an important tool for quality

    management of pharmaceuticals.

    The purpose of this guideline is limited to non-sterile dosage form,

    although the principles and

    Elements specified are equally applicable to sterile products. There are

    various additional

    Guidelines, such as steam sterilization, aseptic processing, radiation

    sterilization, etc., that

    should be considered when the manufacture of sterile products is

    subjected to the process

    validation. This guideline is not intended to specify how validation is tobe conducted, but to

    indicate what the expectations of SFDA from the manufacturers are.

    APPROACH TO PROCESS VALIDATION

    For purposes of this guidance, process validation is defined as thecollection and evaluation of data, from the process design stage through

    commercial production, which establishes scientific evidence that a

    process is capable of consistently delivering quality product.

    guidance describes process validation activities in three stages.

    y Stage 1 Process Design: The commercial manufacturing process is defined during this stage based on knowledge gained

    through development and scale-up activities.

    y Stage 2 Process Qualification: During this stage, the processdesign is evaluated to determine if the process is capable of

    reproducible commercial manufacturing.y Stage 3Continued Process Verification: Ongoing assurance

    is gained during routine production that the process remains in a state

    of control

  • 8/6/2019 ayush reportnew 1 (2)

    6/41

    (6)

    Before any batch from the process is commercially distributed for use

    by consumers, a manufacturer should have gained a high degree of

    assurance in the performance of the manufacturing process such that it

    will consistently produce APIs and drug products meeting those

    attributes relating to identity, strength, quality, purity, and potency.The assurance should be obtained from objective information and data

    from laboratory-, pilot-, and/or commercial-scale studies. Information

    and data should demonstrate that the commercial manufacturing

    process is capable of consistently producing acceptable quality

    products within commercial manufacturing conditions.

    A successful validation program depends upon information and

    knowledge from product and process development. This knowledge

    and understanding is the basis for establishing an approach to control

    of the manufacturing process that results in products with the desired

    quality attributes. Manufacturers should:

    Understand the sources of variation

    Detect the presence and degree of variation

    Control the variation in a manner commensurate with the risk it

    represents to the process and product

    GENERAL CONSIDERATIONS FOR PROCESS

    VALIDATION

    In all stages of the product lifecycle, good project management and good

    archiving that capture scientific knowledge will make the processvalidation program more effective and efficient. The following practices

    should ensure uniform collection and assessment of information about

    the process and enhance the accessibility of such information later in

    the product lifecycle.

  • 8/6/2019 ayush reportnew 1 (2)

    7/41

    (7)

    y We recommend an integrated team approach to process validation that

    includes expertise from a variety of disciplines (e.g., process

    engineering, industrial pharmacy, analytical chemistry, microbiology,

    statistics, manufacturing, and quality assurance). Project plans, along

    with the full support of senior management, are essential elements forsuccess.

    y Throughout the product lifecycle, various studies can be initiated to

    discover, observe, correlate, or confirm information about the product

    and process. All studies should be planned and conducted according

    to sound scientific principles, appropriately documented, and

    approved in accordance with the established procedure appropriate for

    the stage of the lifecycle.

    y The terms attribute(s) (e.g., quality, product, component) andparameter(s) (e.g., process, operating, and equipment) are not

    categorized with respect to criticality in this guidance. With a lifecycle

    approach to process validation that employs risk based decision making

    throughout that lifecycle, the perception of criticality as a continuum

    rather than a binary state is more useful. All attributes and parameters

    should be evaluated in terms of their roles in the process and impact on

    the product or in-process material, and reevaluated as new information

    becomes available. The degree of control over those attributes or parameters should be commensurate with their risk to the process and

    process output. In other words, a higher degree of control is appropriate

    for attributes or parameters that pose a higher risk. The Agency

    recognizes that terminology usage can vary and expects that each

    manufacturer will communicate the meaning and intent of its

    terminology and categorization to the Agency.

    y Many products are single-source or involve complicated

    manufacturing processes. Homogeneity within a batch and

    consistency between batches are goals of process validation activities.Validation offers assurance that a process is reasonably protected

    cause supply problems, and negatively affect public health.

  • 8/6/2019 ayush reportnew 1 (2)

    8/41

    (8)

    TYPES OF PROCESS VALIDATION-

    process validation are divided in 3 category-

    y

    Prospective validationy Concurrent validation

    y Retrospective validation

    PROSPECTIVE VALIDATION-

    In Prospective Validation, the validation protocol is executed before the

    process is put

    into commercial use. During the product development phase the

    production process

    should be broken down into individual steps. Each step should be

    evaluated on the

    basis of experience or theoretical considerations to determine the

    critical parameters

    that may affect the quality of the finished product. A series of

    experiments should be

    designed to determine the criticality of these factors. Each experiment

    should be

    planned and documented fully in an authorized protocol.

    All equipment, production environment and the analytical testing

    methods to be used

    should have been fully validated. Master batch documents can be

    prepared only afterthe critical parameters of the process have been identified and machine

    settings,

  • 8/6/2019 ayush reportnew 1 (2)

    9/41

    (9)

    component specifications and environmental conditions have been

    determined.

    It is generally considered acceptable that three consecutive

    batches/runs within the

    finally agreed parameters, giving product of the desired quality would

    constitute a

    proper validation of the process. It is a confirmation on the commercial

    three batches

    before marketing.

    The matrix approach generally means a plan to conduct process

    validation on

    different strengths of the same product, whereas the family approach

    means a plan

    to conduct process validation on different products manufactured with

    the same

    processes using the same equipment. The validation process using these

    approaches must include batches of different strengths or products

    which should be selected to represent the worst case conditionsor scenarios to demonstrate that the process is consistent for all

    strengths or products

    involved.

  • 8/6/2019 ayush reportnew 1 (2)

    10/41

    (10)

    CONCURRENT VALIDATION-

    Concurrent validation may be the practical approach under certaincircumstances.

    Examples of these may be when:

    A previously validated process is being transferred to a third party

    contract

    manufacturer or to another manufacturing site.

    The product is a different strength of a previously validated product

    with the same

    ratio of active/inactive ingredients

    The number of lots evaluated under the Retrospective Validation were

    not

    sufficient to obtain a high degree of assurance demonstrating that the

    process is

    fully under control.

    The number of batches produced are limited (e.g. orphan drugs).

    Process with low production volume per batch ( e.g.

    radiopharmaceuticals, anticancer).

    Process of manufacturing urgently needed drugs due to shortage (or

    absence) of supply.

    It is important in these cases however, that the systems and equipment

    to be used

    have been fully validated previously. The justification for conducting

    concurrent

  • 8/6/2019 ayush reportnew 1 (2)

    11/41

    (11)

    validation must be documented and the protocol must be approved by

    the Validation

    Team. A report should be prepared and approved prior to the sale of

    each batch and a final report should be prepared and approved after the

    completion of all concurrent batches. It is generally considered acceptable that a

    minimum of three consecutive batches within the finally agreed parameters,

    giving the product the desired quality would constitute a proper

    validation of the process.

    RETROSPECTIVE VALIDATION-

    In many establishments, processes that are stable and in routine use

    have not

    undergone a formally documented validation process. Historical data

    may be utilized

    to provide necessary documentary evidence that the processes are

    validated. The steps involved in this type of validation still require the

    preparation of a protocol,

    the reporting of the results of the data review, leading to a conclusion

    and

    recommendation.

    Retrospective validation is only acceptable for well established detailed

    processes that include operational limits for each critical step of the

    process and will be inappropriate where there have been recent

  • 8/6/2019 ayush reportnew 1 (2)

    12/41

    (12)

    changes in the formulation of the product, operating procedures,

    equipment and facility. The source of data for retrospective validation

    should include amongst others, batch documents, process control

    charts, maintenance log books, process capability studies, finished

    product test results, including trend analyses, and stability results. Forthe purpose of retrospective validation studies, it is considered

    acceptable that data from a minimum of ten consecutive batches

    produced be utilized. When less than ten batches are available, it is

    considered that the data are not sufficient to demonstrate

    retrospectively that the process is fully under control. In such cases the

    study should be supplemented with data generated with concurrent or

    prospective validation.

    Some of the essential elements for Retrospective Validation are:

    y Batches manufactured for a defined period (minimum of 10 last

    consecutive batches).

    y Number of lots released per year.

    Batch size/strength/manufacturer/year/period.

    Master manufacturing/packaging documents.

    Current specifications for active materials/finished products.

    List of process deviations, corrective actions and changes to

    manufacturing documents.

    y Data for stability testing for several batches.

    Trend analyses including those for quality related complaints.

  • 8/6/2019 ayush reportnew 1 (2)

    13/41

    (13)

    PROCESS RE-VALIDATION-

    Re-validation

    becomes necessary in certain situations. The following are examples of

    some of the planned or unplanned changes that may require re-validation:

    Changes in raw materials (physical properties such as density,

    viscosity, particle size distribution, and moisture, etc., that may affect

    the process or product). Changes in the

    source of active raw material manufacturer.

    Changes in packaging material (primary container/closure system).

    Changes in the process (e.g., mixing time, drying temperatures and

    batch size) Changes in the equipment (e.g. additionof automatic detection system). Changes of equipment which involve

    the replacement of equipment on a like for like basis would not

    normally require a re-validation except that this new equipment must

    be qualified. Changes in the plant/facility.

    Variations revealed by trend analysis (e.g. process drifts).

    PRINCIPLE OF PROCESS VALIDATION-Stage 1 Process Design

    Process design is the activity of defining the commercial manufacturing

    process that will be reflected in planned master production and control

    records. The goal of this stage is to design a process suitable for routine

    commercial manufacturing that can consistently deliver a product that

    meets its quality attributes.

    1. Building and Capturing Process Knowledge and Understanding.

  • 8/6/2019 ayush reportnew 1 (2)

    14/41

    (14)

    Generally, early process design experiments do not need to be

    performed under the CGMP conditions required for drugs intended for

    commercial distribution that are manufactured during Stage 2 (process

    qualification) and Stage 3 (continued process verification). They should,

    however, be conducted in accordance with sound scientific methods andprinciples, including good documentation practices. This

    recommendation is consistent with ICH Q10 Pharmaceutical Quality

    System. Decisions and justification of the controls should be sufficiently

    documented and internally reviewed to verify and preserve their value

    for use or adaptation later in the lifecycle of the process and product.

    Although often performed at small-scale

    laboratories, most viral inactivation and impurity clearance studies

    cannot be considered early process design experiments. Viral and

    impurity clearance studies intended to evaluate and estimate product

    quality at commercial scale should have a level of quality unit oversight

    that will ensure that the studies follow sound scientific methods and

    principles and the conclusions are supported by the data.

    Designing an efficient process with an effective process control approach is

    dependent on the process knowledge and understanding obtained. Design of

    Experiment (DOE) studies can help develop process knowledge by revealing

    relationships, including multivariate interactions, between the variable inputs

    (e.g., component characteristics 13 or process parameters) and the resultingoutputs (e.g., in-process material, intermediates, or the final product).

    Risk analysis tools can be used to screen potential variables for DOE studies

    to minimize the total number of experiments conducted while maximizing

    knowledge gained. The results of DOE studies can provide justification for

    establishing ranges of incoming component quality, equipment, parameters,

    and in-process material quality attributes. FDA does not generally expect

    manufacturers to develop and test the process until it fails. It is important to

    understand the degree to which models represent the commercial process,including any differences that might exist, as this may have an impact on the

    relevance of information derived from the models.

  • 8/6/2019 ayush reportnew 1 (2)

    15/41

    (15)

    It is essential that activities and studies resulting in process

    understanding be documented. Documentation should reflect the basis

    for decisions made about the process. For example, manufacturers

    should document the variables studied for a unit operation and the

    rationale for those variables identified as significant. This information isuseful during the process qualification and continued process

    verification stages, including when the design is revised or the strategy

    for control is refined or changed.

    ESTABLISHING A STRATEG Y FOR

    PROCESS CONTROL-

    Process knowledge and understanding is the basis for establishing anapproach to process control for each unit operation and the process overall.

    Strategies for process control can be designed to reduce input variation,

    adjust for input variation during manufacturing (and so reduce its impact on

    the output), or combine both approaches. FDA expects controls to include

    both examination of material quality and equipment monitoring. Special

    attention to control the process through operational limits and in-process

    monitoring is essential in two possible scenarios:

    y When the product attribute is not readily measurable due to

    limitations of sampling or delectability (e.g., viral clearance ormicrobial contamination) or

    y When intermediates and products cannot be highly characterized

    and well-defined quality attributes cannot be identified.

    More advanced strategies, which may involve the use of process analytical

    technology (PAT), can include timely analysis and control loops to adjust

    the processing conditions so that the output remains constant.

    Manufacturing systems of this type can provide a higher degree of process

    control than non-PAT systems. In the case of a strategy using PAT, the

    approach to process qualification will differ from that used in other process

  • 8/6/2019 ayush reportnew 1 (2)

    16/41

  • 8/6/2019 ayush reportnew 1 (2)

    17/41

  • 8/6/2019 ayush reportnew 1 (2)

    18/41

    (18)

    PROCESS PERFORMANCE QUALIFICATION

    The process performance qualification (PPQ) is the second element of

    Stage 2, process qualification. The PPQ combines the actual facility,

    utilities, equipment (each now qualified), and the trained personnelwith the commercial manufacturing process, control procedures, and

    components to produce commercial batches. A successful PPQ will

    confirm the process design and demonstrate that the commercial

    manufacturing process performs as expected. Success at this stage

    signals an important milestone in the product lifecycle. A manufacturer

    must successfully complete PPQ before commencing commercial

    distribution of the drug product.

    The decision to begin commercial distribution should be supported bydata from commercial-scale batches. Data from laboratory and pilot

    studies can provide additional assurance that the commercial

    manufacturing process performs as expected. The cumulative data from

    all relevant studies (e.g., designed experiments; laboratory, pilot, and

    commercial batches) should be used to establish the manufacturing

    conditions in the PPQ. To understand the commercial process

    sufficiently, the manufacturer will need to consider the effects of scale.

    However, it is not typically necessary to explore the entire operatingrange at commercial scale if assurance can be provided by process

    design data. Previous credible experience with sufficiently similar

    products and processes can also be helpful. In addition, we strongly

    recommend firms employ objective measures (e.g., statistical metrics)

    wherever feasible and meaningful to achieve adequate assurance.

    In most cases, PPQ will have a higher level of sampling, additional

    testing, and greater scrutiny of process performance than would be

    typical of routine commercial production. The level of monitoring and

    testing should be sufficient to confirm uniform product quality

    throughout the batch. The increased level of scrutiny, testing, and

    sampling should continue through the process verification stage as

    appropriate, to establish levels and frequency of routine sampling and

  • 8/6/2019 ayush reportnew 1 (2)

    19/41

    (19)

    monitoring for the particular product and process. Considerations for

    the duration of the heightened sampling and monitoring period could

    include, but are not limited to, volume of production, process

    complexity, level of process understanding, and experience with similar

    products and processes.

    The extent to which some materials, such as column resins or molecular

    filtration media, can be re-used without adversely affecting product quality

    can be assessed in relevant laboratory studies. The usable lifetimes of such

    materials should be confirmed by an ongoing PPQ protocol during

    commercial manufacture.

    A manufacturing process that uses PAT may warrant a different PPQ

    approach. PAT processes are designed to measure in real time the

    attributes of an in-process material and then adjust the process in atimely control loop so the process maintains the desired quality of the

    output material. The process design stage and the process qualification

    stage should focus on the measurement system and control loop for the

    measured attribute. Regardless, the goal of validating any

    manufacturing process is the same: to establish scientific evidence that

    the process is reproducible and will consistently deliver quality

    products.

  • 8/6/2019 ayush reportnew 1 (2)

    20/41

    (20)

    PPQ PROTOCOL-

    A written protocol that specifies the manufacturing conditions, controls,

    testing, and expected outcomes is essential for this stage of process

    validation. We recommend that the protocol discuss the followingelements:

    y The manufacturing conditions, including operating parameters,

    processing limits, and component (raw material) inputs.

    y The data to be collected and when and how it will be evaluated.

    y Tests to be performed (in-process, release, characterization) and

    acceptance criteria for each significant processing step.

    y The sampling plan, including sampling points, number of

    samples, and the frequency of sampling for each unit operation

    and attribute. The number of samples should be adequate to

    provide sufficient statistical confidence of quality both within a

    batch and between batches. The confidence level selected can

    be based on risk analysis as it relates to the particular attribute

    under examination. Sampling during this stage should be more

    extensive than is typical during routine production.

  • 8/6/2019 ayush reportnew 1 (2)

    21/41

    (21)

    y Criteria and process performance indicators that allow for a

    science- and risk-based decision about the ability of the process

    to consistently produce quality products. The criteria should

    include:

    y A description of the statistical methods to be used in analyzing

    all collected data (e.g., statistical metrics defining both intra-

    batch and inter-batch variability).

    y Provision for addressing deviations from expected conditions

    and handling of nonconforming data. Data should not be

    excluded from further consideration in terms of PPQ without adocumented, science-based justification.

    y Design of facilities and the qualification of utilities and

    equipment, personnel training and qualification, and

    verification of material sources (components and

    container/closures), if not previously accomplished.

    y Status of the validation of analytical methods used in measuring

    the process, in-process materials, and the product.

    y Review and approval of the protocol by appropriate

    departments and the quality unit.

  • 8/6/2019 ayush reportnew 1 (2)

    22/41

    (22)

    PPQ PROTOCOL EXECUTION AND REPORT-

    Execution of the PPQ protocol should not begin until the protocol has been

    reviewed and approved by all appropriate departments, including the quality

    unit. Any departures from the protocol must be made according toestablished procedure or provisions in the protocol. The PPQ lots should be

    manufactured under normal conditions by the personnel routinely expected

    to perform each step of each unit operation in the process.

    Normal operating conditions should include the utility systems (e.g., air

    handling and water purification), material, personnel, environment, and

    manufacturing procedure.

    A report documenting and assessing adherence to the written PPQ protocol

    should be prepared in a timely manner after the completion of the protocol.This report should:

    y Discuss and cross-reference all aspects of the protocol.

    Summarize data collected and analyze the data, as specified by

    the protocol.

    y Evaluate any unexpected observations and additional data not

    specified in the protocol. Summarize and discuss all

    manufacturing nonconformances such as deviations, aberrant

    test results, or other information that has bearing on the validity

    of the process.

    y Describe in sufficient detail any corrective actions or changes

    that should be made to existing procedures and controls.

    y State a clear conclusion as to whether the data indicates the

    process met the conditions established in the protocol and

    whether the process is considered to be in a state of control. If

    not, the report should state what should be accomplished beforesuch a conclusion can be reached. This conclusion should be

    based on a documented justification for the approval of the

    process, and release of lots produced by it to the market in

    consideration of the entire compilation of knowledge

    .information gained from the design stage.

  • 8/6/2019 ayush reportnew 1 (2)

    23/41

    (23)

    STAGE 3 Continued Process Verification-

    The goal of the third validation stage is continual assurance that the

    process remains in a state of control (the validated state) duringcommercial manufacture. A system or systems for detecting unplanned

    departures from the process as designed is essential to accomplish this

    goal. Adherence to the CGMP requirements, specifically, the collection

    and evaluation of information and data about the performance of the

    process, will allow detection of undesired process variability. Evaluating

    the performance of the process identifies problems and determines

    whether action must be taken to correct, anticipate, and prevent

    problems so that the process remains in control.

    An ongoing program to collect and analyze product and process data

    that relate to product quality must be established. The data collected

    should include relevant process trends and quality of incoming

    materials or components, in-process material, and finished products.

    The data should be statistically trended and reviewed by trained

    personnel. The information collected should verify that the quality

    attributes are being appropriately controlled.

    a statistician or person with adequate training in statistical processcontrol techniques develop the data collection plan and statistical

    methods and procedures used in measuring and evaluating process

    stability and process capability.

    a process is likely to encounter sources of variation that were not

    previously detected or to which the process was not previously

    exposed. Many tools and techniques, some statistical and others more

    qualitative, can be used to detect variation, characterize it, and

    determine the root cause. We recommend that the manufacturer use

    quantitative, statistical methods whenever appropriate and feasible.

    Continued monitoring and sampling of process parameters and quality

    attributes at the level established during the process qualification stage

    until sufficient data are available to generate significant variability

  • 8/6/2019 ayush reportnew 1 (2)

    24/41

    (24)

    estimates. These estimates can provide the basis for establishing levels

    and frequency of routine sampling and monitoring for the particular

    product and process. Monitoring can then be adjusted to a statistically

    appropriate and representative level. Process variability shoul Variation

    can also be detected by the timely assessment of defect complaints, out-of-specification findings, process deviation reports, process yield

    variations, batch records, incoming raw material records, and adverse

    event reports. d be periodically assessed and monitoring adjusted

    accordingly.Data gathered during this stage might suggest ways to

    improve and/or optimize the process by altering some aspect of the

    process or product, such as the operating conditions (ranges and set-

    points), process controls, component, or in-process material

    characteristics. Maintenance of the facility, utilities, and equipment is

    another important aspect of ensuring that a process remains in control.Once established, qualification status must be maintained through

    routine monitoring, maintenance, and calibration procedures and

    schedules. The equipment and facility qualification data should be

    assessed periodically to determine whether re-qualification should be

    performed and the extent of that re-qualification.

    CONCURRENT RELEASE OF PPQ BATCHES-

    In most cases, the PPQ study needs to be completed successfully and a

    high degree of assurance in the process achieved before commercial

    distribution of a product. In special situations, the PPQ protocol can be

    designed to release a PPQ batch for distribution before complete

    execution of the protocol steps and activities, i.e., concurrent release.

    FDA expects that concurrent release will be used rarely.

    Concurrent release might be appropriate for processes used

    infrequently for various reasons, such as to manufacture drugs for

  • 8/6/2019 ayush reportnew 1 (2)

    25/41

    (25)

    which there is limited demand (e.g., orbhan drugs, minor use and minor

    species veterinary drugs) or which have short half lives (e.g.,

    radiopharmaceuticals, including positron emission tomography drugs).

    Concurrent release might also be appropriate for drugs that are

    medically necessary and are being manufactured in coordination withthe Agency to alleviate a short supply.

    Conclusions about a commercial manufacturing process can only be

    made after the PPQ protocol is fully executed and the data are fully

    evaluated. If Stage 2 qualification is not successful then additional

    design studies and qualification may be necessary. Full execution of

    Stages 1 and 2 of process validation is intended to preclude or minimize

    that outcome.

    Circumstances and rationale for concurrent release should be fullydescribed in the PPQ protocol. Even when process performance

    assessment based on the PPQ protocol is still outstanding, any lot

    released concurrently must comply with all CGMPs, regulatory approval

    requirements, and PPQ protocol lot release criteria. Lot release under a

    PPQ protocol is based upon meeting confidence levels appropriate for

    each quality attribute of the drug.

    When warranted and used, concurrent release should be accompanied

    by a system for careful oversight of the distributed batch to facilitaterapid customer feedback. For example, customer complaints and defect

    reports should be rapidly assessed to determine root cause and whether

    the process should be improved or changed. It is important that stability

    test data be promptly evaluated to ensure rapid detection and

    correction of any problems.

  • 8/6/2019 ayush reportnew 1 (2)

    26/41

    (26)

    CHANGE CONTROL-

    Clearly defined procedure are required in order to control any changes

    in the Production processes. These procedures should control all the

    planned changes and ensure the presence of sufficient supporting datathat show that modified process will

    result in a product of the desired quality. Significant changes to process

    (e.g. mixing

    time, drying temperature, etc.), using new equipments with different

    operating

    parameters, etc may require the pre-approval of the SFDA.

    If a change is proposed in any of the procedures, product, processes, or

    equipment,

    which may impact the quality, appropriate written procedures should

    be in place.

    All changes must be formally requested, documented and accepted by

    the Validation

    Team. The proposed changes was scientifically assessed and, depending

    on the

    changes, the need of re-validation will be determined.

    Commitment of the company to control all changes to premises,

    supporting utilities,

    systems, materials, equipment and processes used in the

    fabrication/packaging of

  • 8/6/2019 ayush reportnew 1 (2)

    27/41

    (27)

    pharmaceutical dosage forms is essential to ensure a continued

    validation status of the

    systems concerned. It is essential for a company to control all changes

    to ensure the validity of thecontinued assurance of validation. The

    change control system should ensure that all

    notified or requested changes are satisfactorily investigated,

    documented and

    authorized. Products made by processes subjected to changes should

    not be released

    for sale without full awareness and consideration of the change by the

    Validation

    Team.

    DOCUMENTATION-

    Documentation at each stage of the process validation lifecycle is

    essential for effective communication in complex, lengthy, and

    multidisciplinary projects. Documentation is important so that

    knowledge gained about a product and process is accessible and

    comprehensible to others involved in each stage of the lifecycle.

    Information transparency and accessibility are fundamental tenets of

    the scientific method. They are also essential to enabling organizational

    units responsible and accountable for the process to make informed,

    science-based decisions that ultimately support the release of a product

    to commerce.

  • 8/6/2019 ayush reportnew 1 (2)

    28/41

    (28)

    The degree of documentation is greater for stage 2 (process

    qualification) and stage 3(continued process verification) of the stages

    of process validation as compared to stage 1(process design). All studies

    during these processes should be according the GMP.CGMP documents

    for commercial manufacturing (i.e., the initial commercial master batchproduction and control record and supporting procedures) are key

    outputs of Stage 1, process design.

    Process flow diagrams should describe each unit operation, its

    placement in the overall process, monitoring and control points, and the

    component, as well as other processing material inputs (e.g., processing

    aids) and expected outputs (i.e., in-process materials and finished

    product). It is also useful to generate and preserve process flow

    diagrams of the various scales as the process design progresses to

    facilitate comparison and decision making about their comparability.

    For each stage of the process validation, documentation is very

    essential.

    ANALYTICAL METHODOLOGY-

    Although the validated analytical methods may not be required during

    the product and

    process development activities, the methods used should bescientifically sound (e.g.

    specific, sensitive and accurate), suitable and reliable for the purpose.

    Process knowledge depends on accurate and precise measuring techniques

    used to test and examine the quality of drug components, in-process

    materials, and finished products. Validated analytical methods are not

    necessarily required during product- and process-development activities or

    when used in characterization studies. Nevertheless, analytical methods

  • 8/6/2019 ayush reportnew 1 (2)

    29/41

    (29)

    should be scientifically sound (e.g., specific, sensitive, and accurate) and

    provide results that are reliable.

    There should be assurance of proper equipment function for laboratory

    experiments. Procedures for analytical method and equipment maintenance,

    documentation practices, and calibration practices supporting process-

    development efforts should be documented or described.

    New analytical technology and modifications to existing technology are

    continually being developed and can be used to characterize the process or

    the product. Use of these methods is particularly appropriate when they

    reduce risk by providing greater understanding or control of product quality.

    However, analytical methods supporting commercial batch release must

    follow CGMPs in parts 210 and 211. Clinical supply production should

    follow the CGMPs appropriate for the particular phase of clinical studies.

    VALIDATION PROTOCOL-

    The validation protocol provides a synopsis of what is hoped to be

    accomplished. The protocol should list the selected process and control

    parameters, state the number of batches to be included

    in the study, and specify how the data, once assembled, will be treatedfor relevance. The date of approval by the validation team should also

    be noted. In the case where a protocol is altered or modified after its

    approval, appropriate reasoning for such a change must be documented.

    The validation protocol should be numbered, signed and dated, and

    should contain as a minimum the following information:

    Objectives, scope of coverage of the validation study.

  • 8/6/2019 ayush reportnew 1 (2)

    30/41

    (30)

    Validation team membership, their qualifications and responsibilities.

    Type of validation: prospective, concurrent, retrospective, re-

    validation.

    Number and selection of batches to be on the validation study.

    A list of all equipment to be used; their normal and worst case

    operating parameters.

    Outcome of IQ, OQ for critical equipment.

    Requirements for calibration of all measuring devices.

    Description of the processing steps: copy of the master documents for

    the product.

    Sampling points, stages of sampling, methods of sampling, sampling

    plans.

    Statistical tools to be used in the analysis of data.

    Training requirements for the processing operators.

    Validated test methods to be used in in-process testing and for the

    finished product.

    Specifications for raw and packaging materials and test methods.

    Forms and charts to be used for documenting results.

    Format for presentation of results, documenting conclusions and for

    approval of study

    Result.

  • 8/6/2019 ayush reportnew 1 (2)

    31/41

    (31)

    TEST REPORT-

    When the validation carried out, the actual data is recorded, compared

    with the requirement

    and acceptance criteria(s), and a conclusion is made and authorized in

    form of report.

    Validation Master Plan-

    A validation master plan is a document that summaries the companys

    overall philosophy,intentions and approaches to be used for

    establishing performance adequacy. The Validation Master Plan should

    be agreed upon by management.The validation master plan should

    provide an overview of the entire validation operation, itsorganizational

    structure, its content and planning. The main elements of it being the

    list/inventory of the items to be validated and the planning schedule. All

    validation activities relating to critical technical operations, relevant to

    product and process controls within a firm should be included in the

    validation master plan. It should comprise all prospective, concurrentand retrospective validations as well as re-validation.

    The Validation Master Plan should be a summary document and should

    therefore be brief, concise and clear. It should not repeat information

    documented elsewhere but should refer to existing documents such as

    policy documents, SOPs and validation protocols and reports.

    The format and content should include:

    Introduction: validation policy, scope, location and schedule.

    Organizational structure: personnel responsibilities.

    Plant/process/product description: rational for inclusions or

    exclusions and extent of validation.

  • 8/6/2019 ayush reportnew 1 (2)

    32/41

    (32)

    Specific process considerations that are critical and those requiring

    extra attention.

    List of products/ processes/ systems to be validated, summarized in a

    matrix format, validation approach.

    Re-validation activities, actual status and future planning.

    Key acceptance criteria.

    Documentation format.

    Reference to the required SOPs.

    Time plans of each validation project and sub-project

  • 8/6/2019 ayush reportnew 1 (2)

    33/41

    (33)

    PROCESS VALIDATION DURING

    CLINICAL DEVELOPMENT OF

    BIOLOGICAL MEDICINAL PRODUCTS

    Some critical processes will need to be validated even at phase I.

    These include:

    y Aseptic processing, where possible.

    y viral clearance validation should be no less rigorous than for

    products authorized for marketing.

    y Homogeneity issues.

    y Removal of critical impurities (highly toxic, immunogenic,

    mutagenic,prions, etc.) to levels below the limit of detection, where

    relevant.

    Although for phase I full validation will not be required there are initialsteps towards validation that will need to be taken, such as qualification

    of equipment and of analytical methods. The FDA guidance Sterile Drug

    Products Produced by Aseptic Processing Current Good Manufacturing

    Practice provides for validation requirements for aseptic processes and

    this is expected to be completed as soon as practicable during the initial

    stages of the new drug investigation.

  • 8/6/2019 ayush reportnew 1 (2)

    34/41

    (34)

    LATE PHASE-

    Late phase is generally defined as post-proof-of-concept, which is close

    to the end of phase II and the beginning of phase III. The following is a

    recommended outline of activities related to process validation:

    y end of phase II/phase III: consider whether any critical parts of the

    process should be validated and justify omissions.

    y prior to medical administrive activities/new drug application/BLA.

    y completion of validation.y revalidation following scale-up.

    y manufacture of at least three qualification batches.

    Process validation cannot take place until the equipment, facilities and

    services have been qualified and standard operating procedures

    prepared. stablishing appropriate validation acceptance criteria (VAC)

    is one of the greatest challenges in the development of a commercial

    biological IMP manufacturing process. Manufacturers with VACs that

    are too broad will not be able to demonste adequate process control.

    However, if the manufacturer sets its VAC too tight this can result in

    failed validation runs, even though the process may be performing

    adequately.

    In addition, there is a need to understand potential impurities and have

    validated methods available to detect these. Target limits will need to be

    set and justified for in process control, release and end of shelf-life

    testing. These limits should be based on data amassed during biological

    IMP production and process development as well as trend analyses,

    pharmacopoeial and other regulatory requirements and precedence,

    including assessment of safety.

  • 8/6/2019 ayush reportnew 1 (2)

    35/41

    (35)

    The following issues are often encountered during inspections

    pertaining to process validation and are often quoted in inspection

    reports, such as 483 observations, by investigators:

    y lack of documented procedures and documented validation results.

    y sampling or sample preparation step contributing to overall error.

    y accessories and materials used for equipment qualification not

    qualified.

    y lack of computer system validation (this is in addition to the software

    and hardware qualification requirements).

    y qualification and validation are carried out at just one particular point

    in time.

    y adaptation of acceptance criteria for qualification of new system

    without adequate justification.

    The Process and Its Validation-Validation requirements differ in terms of the specific manufacturing

    process involved and whether the process occurs upstream,

    downstream or fill and finish.

    Upstream-

    Fermentation represents the critical component of the upstream

    process and is typically scaled-up several times, which may impact on

    biological IMP quality, safety and/or efficacy. Thus, to support thescale-

    up activities, a comprehensive physico-chemical and biological testing

    programme will be required. If differences are detected, supporting

    non-clinical and clinical data may be required for late phase changes.

    For phase I, the facilities and equipment will need to have been

    qualified. Trend analyses will continue with full validation being

    conducted on at least three consecutive batches. Upstream

    validation will need to justify in-process controls. For example, this canbe performed by testing the process at the extremities of the limits.

  • 8/6/2019 ayush reportnew 1 (2)

    36/41

    (36)

    DOWNSTREAM-

    Downstream processing involves purification and such critically

    important processes as virus clearance, which are required to be

    validated even at phase I. During later development phases it will alsobe necessary to validate removal of any critical or toxic impurities. By

    medical administrative activities/new drug application/BLA

    submission, validation will need to address issues such as: column

    loading capacity, regeneration, period of use, sanitisation, potential for

    leaching, storage, cleaning and sanitisation. Cleaning/washing and

    sanitisation (percentage of ethanol or sodiumhydroxide) of the column

    packing will be needed to be both established and verified.

    FILL AND FINISH-

    Proteins are generally labile, so terminal sterilisation is usually not

    possible and reliance on aseptic processing is required. This is acritical

    issue that needs to be validated even at phase I. Validation of aseptic

    processes presents special problems when the batch size is small; in

    these cases the number of units filled may be the maximum number

    filled in production. During biological IMP production, filling and sealing

    is often a manual or semi-automated operation presenting great

    challenges to sterility,so enhanced attention should be given to operatortraining and validating the aseptic technique of individual operators.

    Spray drying and lyophilisation are two common processes in the

    finishing of dosage forms. Spray drying involves continuous atomisation

    of the feed solution into a hot drying gas, most commonly air or

    nitrogen. The fine droplets resulting from the atomisation of the feed

    solution are immediately exposed to the drying gas leading to super

    saturation and resulting in the formation of ultra-fine particles, typically

    below 5 and with a tight particle size distribution, which are collectedvia a cyclone. The end product must comply with precise quality

    standards regarding particle size, distribution, residual moisture

    content, bulk density and morphology.

  • 8/6/2019 ayush reportnew 1 (2)

    37/41

    (37)

    Freeze-drying (lyophilisation) has successfully been used for the

    preservation and storage of many vaccines, microbial cultures and other

    labile biological products. Certain biological preparations are

    lyophilised in order to maintain integrity, potency and other properties

    of the product, when for that particular product other methods of

    preservation, such as freezing alone or addition of a preservative, have

    not been found to provide sufficient stability. Residual moisture has

    been the term used to describe the low level of surface water, usually

    from less than 15%, remaining in a freeze-dried vaccine or other

    biological product after the bulk of the aqueous solvent has been

    removed during the freeze-drying (vacuum sublimation) process.

    Examples of freeze-dried biological products include antihemophilic

    factor (human), measles virus vaccine live, streptokinase, Alfa

    interferon, typhoid vaccine, meningococcal polysaccharide vaccine

    groups A and C combined and wasp venom allergenic extract.Over

    drying may cause living cells to lose viability, cause the tertiary

    molecular structure of complex proteins to be altered with subsequent

    loss of activity, or remove monolayers of water from active sites on

    molecules that can then react with traces of oxygen and thus degrade.

    Each product needs to be evaluated on a case-by-case basis to

    determine the optimum residual moisture level. Therefore, the

    approach to process validation should take into considerationdevelopmental data on residual moisture content needed for optimal

    stability of lyophilized products. Some of the common inspectional

    observations pertaining to freeze drying of biopharmaceuticals include

    the following issues:

    y failure to adequately ensure that when the results of a process cannot

    be fully verified by subsequent inspection and test, that the process

    shall be validated with a high degree of assurance and approved

    according to established procedure.

    y Failure to establish acceptance criteria for validation of the freeze-drying process for the manufacturing of a product prior to initiating

    the validation.

    y Assessing or varying different parameters but not evaluating the

    parameters used during the routine freeze-drying process.

  • 8/6/2019 ayush reportnew 1 (2)

    38/41

    (38)

    TYPICAL CONTENT REQUIREMENTS FOR

    PROCESS VALIDATIONS IN BIOLOGICALS-

    It is vital that during all process validation studies, the processes are

    performed in

    the "actual" environment under which production is to occur. That is to

    say all

    routine peripheral activities associated with this process must be in

    effect while the

    validation is being performed. (e.g. number of personnel in facility, exitand entry

    procedures are in effect, environmental and personnel monitoring is

    being performed

    on the prescribed schedule, air system is operating as for regular

    manufacturing.

    CLEANING, FUMIGATION, SANITIZATIONPROCESSES-

    The validation (or re-validation) of these processes includes chemical

    and microbiological testing of samples taken at pre-determined times

    and locations within a facility, a system or piece ofequipment.

    For validation of some cleaning processes, the equipment or surfaces

    can be exposed to an appropriate contaminant (e.g. protein solution,

    microbial strain), the process is performed according to defined

    approved procedures and specifications and then tested to demonstrate

    efficacy.

  • 8/6/2019 ayush reportnew 1 (2)

    39/41

    (39)

    Validation includes collecting liquid and swab samples for testing of

    residual product.

    Typical tests to be performed could include: tests for residual protein,

    endotoxin tests, microbial tests (bioburden), chemical tests (including

    chlorine and phosphoric acid), residual levels of cleaning agents,

    conductivity tests, and pH tests as relevant to the cleaning process

    under test. All analytical tests must be validated before being used in the

    validation of the process. The main considerations in validating a

    cleaning/sanitization/fumigation process are how much of the previous

    active product is left, and how much detergent/cleaning agent remains.

    However there are many tests that should be performed to detect a

    range of different potential contaminants. These include tests for:

    microbial presence, excipient presence, endotoxin contamination,particulate contamination, sanitizing agents, lubricants, environmental

    dust, equipment related contamination and residual rinse water. Worst

    case scenarios should be taken into consideration. For example if any

    residual cleaning agent is distributed unevenly across the test surface,

    then test points must be chosen appropriately.

    STERILIZATION-

    Sterile filtration of solutions: Validation of this process should include a

    microbial challenge that will both test the filter and simulate the

    smallest micro-organism likely to occur in production. Once the filtering

    process is validated it is important to ensure that all replacement filters

    will perform at the same level. This can be done by performing both

    filter integrity tests and performance tests at the same time.

    Equipment: Validation for materials sterilized in the autoclave or oven

    are covered in the Performance qualification.

  • 8/6/2019 ayush reportnew 1 (2)

    40/41

    (40)

    Depyrogenation process-

    The validation (or re-validation) of a depyrogenation (dry heat, column

    chromatography, other) process would include the validation of the

    limits of detection and quantitation of the endotoxin assay, the spikingof samples with endotoxin, running the depyrogenation process

    according to the approved procedures, and the testing of samples for

    residual endotoxin. The full process should be tested at least three times

    to ensure that the process adequately destroys endotoxin and meets the

    required specifications.

    Sterilizing

    Sterile filling tests the filling process for maintenance of asepticconditions by performing the filling process with a nutrient media

    which will easily support bacterial and fungal growth. The filling

    process is run at full scale according to the Master Formula for at least

    one fill size (worst case conditions of large volume and number of vials).

    Facility and system monitoring are performed and recorded during the

    process. The filled vials are incubated, observed and tested for

    contamination by the validated sterility test. The process must be sterile

    for three consecutive runs to be considered validated.

    Typically the media filled container is incubated for 14 days or more at a

    temperature of approximately 25 oC - 35 oC. The media fill is usually

    performed twice a year for each shift for each filling/closing line, but

    this will depend on the frequency required by the regulatory authority.

    The size of the run must be large enough to detect low levels of

    contamination (e.g. for a contamination rate of 1/1000, 3,000 units are

    needed to provide 95% confidence). Appendix 5 includes the validation

    protocol for filling from one of the vaccine manufacturers collaborating

    on the preparation of this guide.

  • 8/6/2019 ayush reportnew 1 (2)

    41/41