a reveiw on pharmaceutical validation

Upload: rajendrashrestha

Post on 11-Oct-2015

262 views

Category:

Documents


14 download

DESCRIPTION

A project file helpful for knowing about pharmaceutical validation

TRANSCRIPT

  • Contents

    Glossary: ....................................................................................................................................................... 5

    1. Introduction to validation .................................................................................................................. 9

    1.1 Definition ............................................................................................................................................ 9

    1.2 History of validation ......................................................................................................................... 10

    1.2.1 Equipment Qualification and Process Qualification: ................................................................. 11

    1.3 Why validation? ................................................................................................................................ 13

    1.4 What has to be validated? ................................................................................................................. 14

    1.5 Scopes of Validation ......................................................................................................................... 15

    1.6 Pre-Requisites for Successful Validation .......................................................................................... 17

    1.7 Approaches of Validation ................................................................................................................. 18

    1.8 Phases in Validation .......................................................................................................................... 18

    1.9 Validation Decision Tree: ................................................................................................................. 20

    2. Organizing for Validation ................................................................................................................ 22

    2.1 Staffing issues ................................................................................................................................... 22

    2.2 Department interactions .................................................................................................................... 22

    2.3 Master planning or planning for Validation ...................................................................................... 24

    2.4 Benefits of Master Planning .............................................................................................................. 24

    2.5 Validation Process ............................................................................................................................ 25

    2.6 Validation Plan .................................................................................................................................. 25

    2.7 Typical validation master plan structure ........................................................................................... 26

    2.8 Validation Protocol ........................................................................................................................... 27

    2.9 Validation set up ............................................................................................................................... 29

    2.10 Relationship between validation and qualification ......................................................................... 29

    2.10.1 Design Qualification (DQ) ....................................................................................................... 30

    2.10.2 Installation Qualification (IQ) .................................................................................................. 32

    2.10.3 Operational Qualification (OQ) ............................................................................................... 33

    2.10.4 Performance Qualification (PQ) .............................................................................................. 34

    2.10.5 Component Qualification (CQ) ................................................................................................ 35

    2.10.6 Requalification ......................................................................................................................... 35

    2.10.7 Revalidation ............................................................................................................................. 35

    2.10.8 Revalidation after change ......................................................................................................... 36

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    2

    2.10.9 Change Control ........................................................................................................................ 36

    2.11 Documentation ................................................................................................................................ 37

    3. Areas of Validation ........................................................................................................................... 43

    3.1 Process Validation ............................................................................................................................ 44

    3.1.1 Pilot Scale-Up and Process Validation ...................................................................................... 45

    3.1.2 Priority Order in Process Validation .......................................................................................... 46

    3.1.3 Stages of Process Validation ...................................................................................................... 47

    3.1.4 Types of process validation ........................................................................................................ 52

    3.1.5 Process Validation Decision ...................................................................................................... 59

    3.1.6 Sterilization Validation .............................................................................................................. 62

    3.2 Analytical method validation ............................................................................................................ 66

    3.2.1 Why analytical methods need to be validated? .......................................................................... 67

    3.2.2 Types of analytical procedures to be validated .......................................................................... 67

    3.2.3 Advantages of analytical method validation .............................................................................. 67

    3.2.3 Strategy for validation of methods ............................................................................................. 68

    3.2.4 Analytical procedure .................................................................................................................. 68

    3.2.5 Validation Parameters ................................................................................................................ 69

    3.2.6 Data Elements Required for Validation ..................................................................................... 76

    3.3 Facilities Validation .......................................................................................................................... 77

    3.3.1 The Engineering Design Process for a Facility .......................................................................... 77

    3.3.2 Conceptual Design: .................................................................................................................... 77

    3.3.3 Purposes: .................................................................................................................................... 78

    3.3.4 Qualification Activities .............................................................................................................. 79

    3.3.5 Qualification Cost ...................................................................................................................... 79

    3.3.6 Design Development: ................................................................................................................. 79

    3.3.7 Facility Qualification Plan ......................................................................................................... 80

    3.3.8 Qualification .............................................................................................................................. 81

    3.4 Computer System Validation ............................................................................................................ 86

    3.4.1 History of computer system validation in brief .......................................................................... 86

    3.4.2 Importance of CSV .................................................................................................................... 87

    3.4.3 Typical Computer System Validation ........................................................................................ 87

    3.4.4 Advantages of CSV .................................................................................................................... 89

    3.4.5 Software validation .................................................................................................................... 90

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    3

    3.4.6 Software Life Cycle ................................................................................................................... 90

    3.4.7 Construction or coding ............................................................................................................... 94

    3.4.8 Testing by the Software Developer ............................................................................................ 94

    3.4.9 User Site Testing ........................................................................................................................ 95

    3.5 Equipment Validation ....................................................................................................................... 96

    3.5.1 Reason of Equipment Validation ............................................................................................... 96

    3.5.2 Content of Equipment Validation .............................................................................................. 96

    3.5.3 Balances and Measuring Equipment .......................................................................................... 97

    3.5.4 Production equipment ................................................................................................................ 97

    3.5.5 Control laboratory equipment .................................................................................................... 97

    3.5.6 Washing, cleaning and drying equipment .................................................................................. 98

    3.5.7 Equipment Validation Process ................................................................................................... 98

    3.5.8 HPLC method calibration ........................................................................................................ 100

    3.5.9 HVAC Validation .................................................................................................................... 107

    3.6 Cold Chain Validation .................................................................................................................... 117

    3.6.1 Uses .......................................................................................................................................... 117

    3.6.2 Strategy .................................................................................................................................... 118

    3.6.3 Evaluation and Reporting......................................................................................................... 119

    3.6.4 Ongoing Monitoring ................................................................................................................ 119

    3.7 Source Validation: .......................................................................................................................... 120

    3.7.1 Methods of vendor validation .................................................................................................. 120

    3.7.2 Corrective and Preventive action ............................................................................................. 123

    3.7.3 Importance of Source Validation ............................................................................................. 124

    3.8 Personnel Validation ....................................................................................................................... 127

    3.8.1 GMP Requirement ................................................................................................................... 127

    3.8.2 Responsibilities ........................................................................................................................ 127

    3.8.3 Training for personnel .............................................................................................................. 129

    3.9 Packaging Validation: ..................................................................................................................... 130

    3.9.1 Packaging Materials ................................................................................................................. 131

    3.9.2 Packaging Equipment .............................................................................................................. 131

    3.9.3 Assess the GMP Risk ............................................................................................................... 132

    3.9.4 Line Layout .............................................................................................................................. 132

    3.9.5 Operating Procedure and Training ........................................................................................... 132

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    4

    3.9.6 Conduct of Packaging Validation ............................................................................................ 133

    3.9.7 Performance qualification examples ........................................................................................ 135

    3.9.8 Tests that can be performed for packaging validation ............................................................. 136

    3.10 Cleaning Validation ...................................................................................................................... 139

    3.10.1 Necessity ................................................................................................................................ 140

    3.10.2 Advantages ............................................................................................................................. 140

    3.10.3 Contamination ........................................................................................................................ 140

    3.10.4 Cross Contamination .............................................................................................................. 140

    3.10.5 Mechanism of Contamination ................................................................................................ 141

    3.10.6 Cleaning Agent selection ....................................................................................................... 141

    3.10.7 Sampling Techniques ............................................................................................................. 142

    3.10.8 Sampling Methods ................................................................................................................. 143

    3.10.9 Level of Cleaning ................................................................................................................... 145

    3.10.10 Cleaning Validation procedure ............................................................................................ 146

    3.10.11 Strategy on Cleaning Validation Studies ............................................................................. 146

    3.10.12 Analyzing cleaning validation samples ................................................................................ 148

    3.10.13 Data analysis for estimating possible contamination ........................................................... 149

    4. Future Aspects of Validation ............................................................................................................. 150

    4.1. Latest Technology .......................................................................................................................... 150

    4.2 Automated Inspection/Identification ............................................................................................... 150

    4.3 Process Automation ........................................................................................................................ 150

    4.4 Robotics .......................................................................................................................................... 151

    4.5 Isolation........................................................................................................................................... 151

    5. Conclusion ....................................................................................................................................... 153

    6. References ........................................................................................................................................ 154

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    5

    Glossary:

    1. Calibration The set of operations that establish, under specified conditions, the

    relationship between values indicated by an instrument or system for measuring (for e.g.

    weight, temperature, pH), recording and controlling, or the values represented by a material

    measure, and the corresponding known values of a reference standard. Limits for acceptance

    of the results of measuring should be established.

    2. Computer validation - Documented evidence which provides a high degree of assurance

    that a computerized system analyses, controls and records data correctly and that data

    processing complies with predetermined specifications.

    3. Commissioning The setting up, adjustment and testing of equipment or a system to ensure

    that it meets all the requirements, as specified in the user requirement specification, and

    capacities as specified by the designer or developer. Commissioning is carried out before

    qualification and validation.

    4. Concurrent validation Validation carried out during routine production of products

    intended for sale.

    5. Cleaning validation Documented evidence to establish that cleaning procedures are

    removing residues to predetermined levels of acceptability, taking into consideration factors

    such as batch size, dosing, toxicology, and equipment size.

    6. Design qualification (DQ) Documented evidence that the premises, supporting systems,

    utilities, equipment and processes have been designed in accordance with the requirements of

    GMP.

    7. Good engineering practices (GEP) Established engineering methods and standards that

    are applied throughout the project life-cycle to deliver appropriate, cost-effective solutions.

    8. Installation qualification (IQ) The performance of tests to ensure that the installations

    (such as machines, measuring devices, utilities and manufacturing areas) used in a

    manufacturing process are appropriately selected and correctly installed and operate in

    accordance with established specifications.

    9. Operational qualification (OQ) Documented verification that the system or subsystem

    performs as intended over all anticipated operating ranges.

    10. Performance qualification (PQ) Documented verification that the equipment or system

    operates consistently and gives reproducibility with defined specifications and parameters for

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    6

    prolonged periods. (In the context of systems, the term process validation may also be

    used.)

    11. Process validation Documented evidence which provides a high degree of assurance that a

    specific process will consistently result in a product that meets its predetermined

    specifications and quality characteristics.

    12. Prospective validation Validation carried out during the development stage on the basis of

    a risk analysis of the production process, which is broken down into individual steps; these

    are then evaluated on the basis of past experience to determine whether they may lead to

    critical situations.

    13. Qualification Action of proving and documenting that any premises, systems and

    equipment are properly installed, and/or work correctly and lead to the expected result.

    Qualification is often a part (the initial stage) of validation, but the individual qualification

    steps alone do not constitute process validation.

    14. Retrospective validation Involves the evaluation of past experience of production on the

    condition that composition, procedures, and equipment remain unchanged.

    15. Revalidation Repeated validation of an approved process (or a part thereof) to ensure

    continued compliance with established requirements.

    16. Standard Operating Procedure (SOP) An authorized written procedure giving

    instructions for performing operations not necessarily specific to a given product or material

    but of a more general nature (e.g. equipment operation, maintenance and cleaning;

    validation; cleaning of premises and environmental control, sampling and inspection).

    Certain SOPs may be used to supplement product-specific master batch production

    documentation.

    17. Validation Action of proving and documenting that any process, procedure or method

    actually and consistently leads to the expected results. The aim of validation is not to correct

    or detect deviations in the packed product but to prevent deviations in the final packed

    products as far as is practicable and economic.

    18. Validation protocol (or plan) (VP) A document describing the activities to be performed

    in a validation, including the acceptance criteria for the approval of a manufacturing process

    or a part thereof for routine use.

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    7

    19. Validation report (VR) A document in which the records, results and evaluation of a

    completed validation programme are assembled and summarized. It may also contain

    proposals for the improvement of processes and/or equipment.

    20. Validation master plan (VMP) - The VMP is a high-level document that establishes an

    umbrella validation plan for the entire project and summarizes the manufacturers overall

    philosophy and approach, to be used for establishing performance adequacy. It provides

    information on the manufacturers validation work programme and defines details of and

    timescales for the validation work to be performed, including a statement of the

    responsibilities of those implementing the plan.

    21. Verification The application of methods, procedures, tests and other evaluations, in

    addition to monitoring, to determine compliance with the GMP principles.

    22. Worst case A condition or set of conditions encompassing the upper and lower processing

    limits for operating parameters and circumstances, within SOPs, which pose the greatest

    chance of product or process failure when compared to ideal conditions. Such conditions do

    not necessarily include product or process failure.

    23. URS User Requirements Specification (URS) provides a clear and precise definition of

    what the user wants the system to do. It defines the functions to be carried out, the data on

    which the system will operate and the operating environment. The URS define also any non-

    functional requirements, constraints such as time and costs and what deliverables are to be

    supplied. The emphasis should be on the required functions and not the method of

    implementing those functions.

    24. Acceptance Criteria The criteria a product must meet to successfully complete a test

    phase or to achieve delivery requirements.

    25. Change Control A formal system of reviewing and documenting proposed or actual

    change that might affect the validated status of a system, equipment or process followed by

    action to ensure ongoing validated state.

    26. Requirement It can be any need or expectation for a system. It reflects the stated or

    implied needs of the customer, and may be market-based, contractual, or statutory, as well as

    an organizations internal needs.

    27. Specification It is a document that states requirements.

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    8

    28. Quality Assurance It can be defined as the totality of the arrangements made with the

    object of ensuring that pharmaceutical products are of the quality required for their intended

    use. In addition, it ensures that arrangements made for the manufacture, supply and use of the

    correct starting and packaging materials.

    29. Quality Control It is the part of GMP concerned with sampling, specifications and testing,

    and with the organization, documentation and release procedures which ensures that the

    necessary and relevant tests are actually carried out and that materials are not released for

    used, nor products released for sale or supply, until their quality has been judged to be

    satisfactory. It is not confined to laboratory operations but must be involved in all decisions

    concerning the quality of the product.

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    9

    1. Introduction to validation

    1.1 Definition

    Validation is not a one-time event but on-going process covering all phases of a product or

    process. Literally, validation in pharmaceuticals means to be valid or justifiable. Simply saying,

    validation means action of proving effectiveness. According to FDA 1987 validation is

    establishing documented evidence which provides a high degree of assurance that a specific

    process will consistently produce a product meeting its predetermined specifications and quality

    attributes.1 According to European Commission- 1991, validation is an act of proving in

    accordance of GMPs that any process actually leads to expected results. According to European

    Commission-2000, validation is documented evidence that the process, operated within

    established parameters, can perform effectively and reproducibly to produce a medicinal product

    meeting its predetermined specifications and quality attributes.

    Validation is the evaluating of processes, products or analytical methods to ensure compliance

    with product or method requirements. Prerequisites to fulfill these requirements for analytical

    laboratories are properly functioning and well documented instruments (hardware and firmware),

    computer hardware and software and validated analytical methods.2

    Validation is an essential part of good manufacturing practices (GMP). It is, therefore, an

    element of the quality assurance programme associated with a particular product or process. The

    basic principles of quality assurance have as their goal the production of products that are fit for

    their intended use. These principles are as follows:

    Quality, safety and efficacy must be designed and built into the product.

    Quality cannot be inspected or tested into the product.

    Each critical step of the manufacturing process must be validated. Other steps in the process

    must be under control to maximize the probability that the finished product consistently and

    predictably meets all quality and design specifications.

    Validation of processes and systems is fundamental to achieving these goals. It is by design and

    validation that a manufacturer can establish confidence that the manufactured products will

    consistently meet their product specifications.

    Documentation associated with validation includes:

    Standard operating procedures (SOPs)

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    10

    Specifications

    Validation master plan (VMP)

    Qualification protocols and reports

    Validation protocols and reports.

    The implementation of validation work requires considerable resources such as:

    Time: Generally validation work is subject to rigorous time schedules.

    Financial: Validation often requires the time of specialized personnel and expensive

    technology.

    Human: Validation requires the collaboration of experts from various disciplines (e.g. a

    multidisciplinary team, comprising quality assurance, engineering, manufacturing and other

    disciplines, depending on the product and process to be validated).3

    Chapman purported Validation means nothing else than well-organized, well-documented

    common sense.4

    1.2 History of validation:

    Validation is a subject that has grown in importance within the global healthcare industry over

    the past 25 years. Its origin can be traced to terminal sterilization process failures in the early

    1970s. Individuals in the US point to the LVP sterilization problems of Abbott and Baxter, while

    those in the U.K. cite the Davenport incident.5 Each incident was a result of a non-obvious fault

    coupled with the inherent limitations of the end-product sterility test. As a consequence of these

    events, non-sterile materials were released to the market, deaths occurred, and regulatory

    investigations were launched. The outcome of this was the introduction by the regulators of the

    concept of Validation.

    The initial reaction to this regulatory initiative was one of puzzlement, only a limited number of

    firms had encountered difficulties, and all of the problems were seemingly associated with the

    sterilization of LVP containers. It took several years for firms across the industry to understand

    that the concerns related to process effectiveness were not limited to LVP solutions, and even

    longer to recognize that those concerns were not restricted to sterile products. From its earliest

    days, validation was identified as a new regulatory requirement to be added to the list of things

    that firms must do, with little consideration of its real implications. The first efforts reflected

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    11

    what can be termed the scientific method of observation of an activity, hypothesis/predition of

    cause/effect relationship, and experimentation followed by new observations in the form of the

    experimental report. In the pharmaceutical validation model this has evolved into the validation

    protocol (hypothesis and prediction), field execution (experimentation), and summary report

    preparation (documented observations).6

    By 1980, it was evident to all that validation was here to stay, so pharmaceutical firms began to

    organize their activities more formally. Ad hoc teams and task forces that had started the efforts

    were replaced by permanent Validation Departments whose responsibilities and scope varied

    with the organization but whose purpose was to provide the necessary validation for a firms

    products and processes. The individuals in these departments were the first to grapple with

    validation as their primary responsibility, and their methods, concepts, and practices have served

    to define validation ever since as establishing documented evidence which provides a high

    degree of assurance that a specific process will consistently produce a product meeting its pre-

    determined specifications and quality attributes.7

    The first efforts at validation were rather crude and limited in their understanding of the full

    implications but slowly made significant strides. For e.g. the first sterilization validations were

    performed without prior qualification of the equipment. Once validation had been established as

    discipline, methods for its execution became substantially more formalized and rigorous.

    Perhaps, most important was stride was separation of activities into two major categories.

    1.2.1 Equipment Qualification and Process Qualification:

    It was apparent by then that validation had to be more closely integrated into the mainstream of

    cGMP operations in order to maximize its effectiveness in larger organizations. A number of

    areas can be identified as pre-requisites for process or system validation. The origins of these

    elements can be identified in the cGMP requirements for drugs and devices (Table 1).8

    With this understanding, the industry began to recognize that validation offered advantages to the

    firm and implemented validation objectives that were non-regulatory and geared for the

    optimization of processes and systems. The attention being placed on validation at this time led

    to important changes in how firms approached its implementation and should be integrated with

    other GMP.

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    12

    Table 1:- Pre-Requisites for Validation

    1 Process Development [21 CFR 820.30Design Control]. The activities performed to

    define the process, product or system to be evaluated.

    2 Process Documentation [21 CFR 211 Subparts F - Production and Process controls and J-

    Records and Reports]. The documentation (batch records, procedures, test methods,

    sampling plans) and processes (software) that define the operation of the equipment to

    attain the desired result.

    3 Equipment Qualification [21CFR 211 Subparts C Buildings and Facilities and D-

    Equipment]. The specifications, drawing, checklists and other data that support the

    physical equipment (hardware) utilized for the process.

    4 Calibration [21 CFR 211 Subparts D Equipment]. The methods and controls that

    establish the accuracy of data.

    5 Analytical Methods [21CFR 211 Subparts I Laboratory Controls]. The means to

    evaluate the outcome of the process on the materials.

    6 Cleaning [21 CFR 211.67Equipment Cleaning and Maintenance]. A specialized process,

    the intent of which is to remove the traces of the prior product from the equipment.

    7 Change Control [21 CFR 211. 100(b) Equipment Cleaning and Maintenance]. A

    formalized process control scheme that evaluates changes to documentation, materials,

    and equipment.

    The pharmaceutical industry participated in the introduction of computers into the manufacturing

    environment during the 1980s. This led to FDA concerns relative to the validation of

    computerized system used within the industry. The pharmaceutical industrys response to the

    FDAs new concerns regarding validation of computerized systems was somewhat different than

    what had occurred previously. The Pharmaceutical Manufacturers Association established an

    interdisciplinary group called the Computer Systems Validation Committee (CSVC) in late 1983

    to address how the industry would address the FDAs concerns. Through the creation of the

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    13

    CSVC, the industry began to assume a position of leadership regarding validation. Through the

    auspices of the CSVC, an industry approach to the validation of computerized systems in the

    GMP environment was established.9 Central to the industry position, was the adoption of the

    life cycle concept as an appropriate model for managing the activities needed for the

    successful validation of computerized systems (Figure 1). The life cycle approach focuses on

    managing a project from cradle to grave. When employing the life cycle approach, the design,

    implementation, and operation of system (or project) are recognized as interdependent parts of

    the whole. Operation and maintenance concerns are addressed during the design of the system

    and confirmed in the implementation phase to ensure their acceptability. The adoption of the life

    cycle methodology afforded such a degree of control over the complex tasks associated with the

    validation of computerized systems that it came into nearly universal application within a very

    short period.

    Figure 1

    1.3 Why validation?

    First and foremost, among the reasons for validation is that it is a regulatory requirement for

    virtually every process in the global healthcare industry for pharmaceuticals, biologics, and

    medical devices. Regulatory agencies across the world expect firms to validate their processes.

    The continuing trend toward harmonization of requirements will eventually result in a common

    level of expectation for validations worldwide.

    Number of tangible and intangible benefits of validation was realized (Table 2)10

    . In the

    intervening years, there has been repeated affirmation of those expectations at other firms, large

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    14

    and small. Regrettably, there has been little quantification of these benefits. The predominance of

    compliance-based validation initiatives generally restricts objective discussion of cost

    implications for any initiative. But once a process/product is properly validated, it seem that

    reduced sample size and intervals could be easily justified, and thus provide a measurable return

    on the validation effort. Aside from utility systems, it is hardly ever realized and represents one

    of the major failings relative to the implementation of validation in pharmaceutical industry.

    Table 1: Benefits of Validation

    Increased throughput

    Reduction in rejections and reworks

    Reduction in utility costs

    Avoidance of capital expenditures

    Fewer complaints about process related failures

    Reduced testing in process and finished goods

    More rapid and accurate investigations into process deviations

    More rapid and reliable startup of new equipment

    Easier scale-up from development work

    Easier maintenance of the equipment

    Improved employee awareness of processes

    More rapid automation

    Validation and validation-like activities are found in a number of industries, regulated and

    unregulated. Banking, aviation, software, microelectronics, nuclear power, among others all

    incorporate practices closely resembling validation of health care product production. The health

    care industries fixation on compliance has perhaps blinded us the real value of validation

    practices.

    1.4 What has to be validated?

    Validation efforts in the analytical laboratory should be broken down into separate components

    addressing the equipment (both the instrument and the computer controlling it) and the analytical

    methods run on that equipment. After these have been verified separately they should be checked

    together to confirm expected performance limits (so-called system suitability testing), and finally

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    15

    the sample analysis data collected on such a system should be authenticated with suitable

    validation checkouts. Other activities include checking reference standards and qualification of

    people.

    1) Equipment: All (computerized) equipment that is used to create, modify, maintain, archive,

    retrieve, or distribute critical data for cGMP/GCP/GLP purposes should be validated.

    Validation of hardware includes testing the instrument according to the documented

    specifications. Even though this may include word processing systems to create and maintain

    SOPs, it covers analytic systems only. If instruments consist of several modules, a modular

    HPLC system for example, the entire system should be validated. Validation of computer

    systems must include the qualification of hardware and software.

    2) Analysis method: Validation covers testing of significant method characteristics, for e.g

    sensitivity and reproducibility.

    3) Analytical system: The system combines instrument, computer and analytical method. This

    validation usually referred to as system suitability testing, tests the system for documented

    performance specifications for the specific analysis method.

    4) Data: When analyzing samples the data must be validated. The validation process includes

    documentation and checks for data plausibility, consistency, integrity, and traceability. A

    complete audit trail must be in place, which allows tracing back the final result to the raw

    data for integrity.

    5) Personnel: People should be qualified for their jobs. This includes education, training and/or

    experience.

    6) Reference standards: Reference standard should be checked for purity, identity,

    concentrations and stability.11

    1.5 Scopes of Validation

    There should be an appropriate and sufficient system including organizational structure and

    documentation infrastructure, sufficient personnel and financial resources to perform

    validation tasks in a timely manner. Management and persons responsible for quality

    assurance should be involved.

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    16

    Personnel with appropriate qualifications and experience should be responsible for

    performing validation. They should represent different departments depending on the

    validation work to be performed.

    There should be proper preparation and planning before validation is performed. There

    should be a specific programme for validation activities.

    Validation should be performed in a structured way according to the documented procedures

    and protocols.

    Validation should be performed for:

    For new premises, equipment, utilities and systems, and processes and procedures.

    At periodic intervals, and

    When major changes have been made.

    (Periodic revalidation or periodic requalification may be substituted, where appropriate, with

    periodic evaluation of data and information to establish whether requalification or revalidation is

    required).

    Validation should be performed in accordance with written protocols. A written report on the

    outcome of the validation should be produced.

    Validation should be done over a period of time, e.g. at least three consecutive batches (full

    production scale) should be validated, to demonstrate consistency. Worst case situations

    should be considered.

    There should be a clear distinction between in-process controls and validation. In-process

    tests are performed during the manufacture of each batch according to specifications and

    methods devised during the development phase. Their objective is to monitor the process

    continuously.

    When a new manufacturing formula or method is adopted, steps should be taken to

    demonstrate its suitability for routine processing. The defined process, using the materials

    and equipment specified, should be shown to result in the consistent yield of a product of the

    required quality.

    Manufacturers should identify what validation work is needed to prove that critical aspects of

    their operations are appropriately controlled. Significant changes to the facilities or the

    equipment, and processes that may affect the quality of the product should be validated. A

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    17

    risk assessment approach should be used to determine the scope and extent of validation

    required.12

    1.6 Pre-Requisites for Successful Validation

    There are some elements or tools that are required for conducting effective validations. Each are

    presented and discussed in the following sections:

    Understanding

    The single most important element required is a good understanding of what validation is.

    This understanding activity goes beyond the basic definition of validation, beyond the

    concept of requiring a minimum of three runs and understanding must be anchored by

    sufficient years of practical experience and knowledge. It will permit sound and logical

    decisions even under most intense situations.13

    Communication

    Communication is one of the best methods of improving environment understanding. It is

    essential for any activity that requires more than one resource to complete. This point is

    understandable considering that conducting effective validation involves multi-departments.

    Co-operation and Focus

    Multi departments that sometimes interact during the course of executing validation program

    are project management, accounting, quality control, project engineering, process

    engineering, quality assurance, facilities; regulatory etc should have a commendable co-

    operation.

    Experience

    A firm must have resources with solid validation experience to get success in their validation

    program.

    Resources

    Resources mean personnel who will plan and execute equipment on which validations will be

    performed on materials with which to conduct validations. Laboratories that will perform

    necessary analysis should provide necessary funding for the validations and allocate

    sufficient time to perform validations.14

    Plan

    Conducting validations within most companies will involve a number of departments and

    disciplines. These disciplines need a perfect plan in order to get good team synergy.

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    18

    Budget

    It is important to understand that a successful validation must be done to completion and it

    should not be limited by a budget assembled by personnel who have no appreciation for what

    is required to successfully complete validation. Further, it is important to understand that

    validations cost money.15

    Standard Operating Procedures (SOPs)

    The SOPs capture activities that routinely occur within an organization. Departments charged

    with abiding by or following these SOPs must first be trained against these SOPs.

    Quality Control lab support

    In most of the validations, some laboratory testing will be required. In most cases this testing

    is handled by the QC group. QC is expected to provide results in timely manner. So often, the

    wait for the receipt of analytical results cases the entire validation project to come to halt.

    Because validations are based on the results obtained.

    1.7 Approaches of Validation

    According to the WHO, there are two basic approaches to validation; one is based on evidence

    obtained through testing (prospective and concurrent validation), and another is based on the

    analysis of accumulated (historical) data (i.e. retrospective validation). Whenever possible,

    prospective validation is preferred. Retrospective validation is no longer encouraged and is, in

    any case, not applicable to the manufacturing of sterile products.

    Both prospective and concurrent validation may include following:

    Extensive product testing, which may involve extensive sample testing (with the estimation

    of confidence limits for individual results) and the demonstration of intra- and inter-batch

    homogeneity.

    Simulation process trials

    Challenge/ worst case tests, which determine the robustness of the process, and

    Control of process parameters being monitored during normal production runs to obtain

    additional information on the reliability of the process.16

    1.8 Phases in Validation17:

    The activities relating to validation studies may be classified into three phases mainly. They are

    as follows:

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    19

    1. Pre-validation Qualification Phase: It covers all activities related to product research and

    development, formulating pilot batch studies, scale-up studies, technology transfer to

    commercial scale batches, establishing stability conditions and storage, and handling of in-

    process and finished dosage forms, equipment; operational and installation qualification,

    master production document and process capacity.

    2. Process validation phase: It is designed to verify that all established limits of the critical

    process parameters are valid and satisfactory products can be produced even under worst

    conditions.

    3. Validation Maintenance phase: It requires frequent review of all process related documents,

    including audit reports, to assure that there have been no changes, deviations, failures and

    modifications to the production process and that all SOPs, including change control

    procedures have been followed. At this phase, the validation comprising of members from all

    major departments assures that there have been no changes/deviations that should be resulted

    in requalification and revalidation. A careful design and validation of systems and process

    controls can establish a high degree of confidence that all lots of batches produced will meet

    their intended specifications. Thus, its assumed that throughout manufacturing and control,

    operations are conducted in accordance with the principle of GMP both in general and in

    specific reference to sterile product manufacture.

    The validation steps recommended in GMP guidelines can be summarized as follows18

    :

    As a pre-requisite, all studies should be conducted in accordance with a detailed, pre-

    established protocol or series of protocols, which in turn is subject to formal change control

    procedures.

    Both the personnel conducting the studies and those running the process being studied should

    be appropriately trained and qualified and be suitable and competent to perform the task

    assigned to them.

    All data generated during the course of studies should be formally reviewed and certified as

    evaluated against pre-determined criteria.

    Suitable testing facilities, equipment, instruments and methodology should be available.

    Suitable clean room facilities should be available in both the local and background

    environment. There should be assurance that the clean room environment as specified is

    secured through initial commissioning (qualification) and subsequently through the

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    20

    implementation of a programme of re-testing in-process equipment should be properly

    installed, qualified and maintained.

    When appropriate attention is paid to above, the process, if aseptic, may be validated by

    means of process simulation studies.

    The process should be revalidated at specific time intervals.

    Comprehensive documentation should be available to define support and record the overall

    validation process.

    1.9 Validation Decision Tree:

    This model describes a decision tree that helps manufacturer decide on whether processes need

    to be validated or not. It is one of the easiest models under consideration.

    Each process should have a specification describing both the process parameters and the desired

    output. The tree is described below:

    A

    Is process

    Output

    Verifiable

    B

    Is Verification

    Sufficient &

    Cost Effective

    C

    Verify &

    Control the

    Process

    D

    Validate

    E

    Redesign

    Product and/or

    Process

    NO NO

    Yes YES

    Figure- 2: Validation Decision Tree

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    21

    A. The manufacturer should consider whether the output can be verified by subsequent

    monitoring or measurement.

    B. If the answer is positive, then the consideration should be made as to whether or not

    verification alone is sufficient to eliminate unacceptable risk and is a cost effective solution.

    C. If yes, the output should be verified and the process should be appropriately controlled.

    D. If the output of the process is not verifiable then the decision should be to validate the

    process.

    E. The product or process should be redesigned to reduce variation, improve product or process

    and decrease risk or cost to a point where verification is acceptable decision.

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    22

    2. Organizing for Validation

    Validation and its role within a pharmaceutical organization have come a long way from its

    inception in the 1970s, when the effort was primarily focused on sterilization validation and

    demonstrating that the conditions to achieve sterility were met. As a result, it was managed from

    within the sterile manufacturing unit using a small team. In the 1980s, validations organizations

    were created and began interacting with other groups such as Research, Engineering, Production,

    Manufacturing, and Quality Assurance.

    Formulating a mission is essential to ensure proper definition of a department role in the

    formation. Although there is broad diversity of validation department missions within the

    pharmaceutical industry, the mission that is general to all validation departments is the satisfying

    of the regulatory requirement to have processes validated. Certainly the validation mission is

    influenced by the size of the company as well as its product lines.

    2.1 Staffing issues

    When staffing a validation group, the mission and the organization exert a degree of influence,

    primarily in the academic backgrounds of the members. Because of the aforementioned diversity,

    a considerable variety of academic backgrounds are usually found among validation

    professionals, such as members having degrees in chemistry, microbiology, pharmacy, statistics,

    computer science, biochemistry as well as engineering disciplines. For e.g. when the mission is

    directed toward a sterile products focus, having a microbiology degree would be beneficial. In

    general sense, the more important than the actual academic background are these 3 skills:

    problem-solving capability, interpersonal skills, and oral and written communication abilities.

    The technical talent to recognize and solve problems is fundamental to validation. Finally, it is

    targeted that the validation members be able to effectively express the validation objectives and

    concerns both orally and in written form. If the professional can successfully communicate

    orally, esp. during an FDA visit, the strength of validation package is expected to be even

    greater.

    2.2 Department interactions

    Once missions of departments have been formalized and the validation operations are organized,

    the main challenge is to implement the plan, which requires interaction with many peer groups.

    Within the company, other departments involved in validation taskforce are as follows:

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    23

    1. R & D: It is involved with new product and process development and often existing process

    improvements. Their key responsibility in validation is to ensure the acceptability (and thus

    validatability) of new products or improved process in the manufacturing area. They must

    be aware of the validation plan and resulting acceptance criteria.

    2. Engineering: It is involved with new or modified equipment or facilities and their start-ups.

    Their role is to ensure the acceptability of the processes later on, so their concern must be

    built in at the design phase and continued through construction.

    3. Production: It is concerned with processes that require validation and stress the benefits of a

    validation program.

    4. Maintenance: Its concerned with change control, calibration, and preventative maintenance.

    This occurs at the instant when an undocumented change is made to a validated piece of

    equipment.

    5. Quality Control: Its involved with the testing laboratories and ensures that laboratory

    personnel know not only the number and type of tests required for the study but also how the

    testing fits into the overall validation program.

    6. Quality Assurance: Its concerned with GMP compliance to ensure a firms regulatory

    compliance. Through the technical competency of the validation staff and the GMP

    compliance expertise existing within the QA group, these efforts should be successful. The

    key point is to communicate so that the regulatory compliance objective of validation is met.

    Whether the process is a bulk process or one of the finishing steps; whether it is a proprietary

    purification process, a steam sterilization process, or a conventional non-sterile process; whether

    the focus is a clinical manufacturing lot or commercial production; or whether the effort is

    accomplished within the firm, contracted out in conjunction with an outsourced manufacturing

    agreement, or with the assistance of a consultant, the validation staff must possess 4 things:

    i. Technical expertise, allowing a thorough understanding of the process being reviewed.

    ii. Understanding of the fundamentals of validation and the ability to apply them to the process.

    iii. Interpersonal skills necessary to deal with all of the organizations within and outside of the

    firm.

    iv. Support from management, which positions the validation effort as a critical element in the

    companys success.

    These basics ensure that the validation effort is successfully accomplished.19

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    24

    2.3 Master planning or planning for Validation

    The validation master plan (VMP) has become common practice for all large capital projects

    within the global healthcare industry. The master plan has come into vogue to ensure that the

    validation requirements for major facilities are adequately addressed. Although its often

    described as a regulatory requirement, there is in fact no such requirement in any of the worlds

    cGMP regulations; nevertheless, its real value is as a management tool to be used to coordinate

    the validation effort. It is an indispensable tool that delineates how the validation effort is to be

    executed. The utility of plan diminishes with facility size and complexity, but even small projects

    may benefit from the structure that a master plan brings to the validation effort.20

    VMP is a good practice to document all validation activities in a document. The FDA does not

    specifically demand a validation master plan however; inspectors want to know what the

    companys approach towards validation is. So, VMP is an ideal tool to communicate this

    approach internally and to inspectors.21

    The validation master plan should provide an overview

    of the entire validation operation, its organizational structure, its content and planning. All

    validation activities relating to critical technical operations, relevant to product and process

    controls within a firm should be included in the VMP. It should comprise all prospective,

    concurrent and retrospective validations as well as revalidation.22

    The VMP should reflect the

    key elements of the validation programme. It should be concise and clear and contain at least the

    following:

    A validation policy

    Organizational structure of validation activities

    Summary of facilities, systems, equipment and processes validated and to be validated.

    Documentation format (e.g. protocol and report format)

    Planning and scheduling

    Change control

    References to existing documents

    2.4 Benefits of Master Planning

    Numerous benefits are derived from a VMP which can substantially enhance the firms

    validation posture for the project. A well-structured plan will provide following advantages to a

    firm:

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    25

    i. Codify decisions regarding how cGMP requirements will be satisfied.

    ii. Allow detailed definition of validation activities necessary for the successful operation of the

    facility.

    iii. Serve as an important document in regulatory compliance and interaction.

    iv. Serve as a communication document on the validation for use with third parties.

    v. Be easily converted into a Drug Master File

    vi. Serve as an excellent tool for audit preparation (either internal or external).

    vii. Define project execution through the definition of requirements.

    viii. Help determine resource needs for personnel, materials, equipment, components and

    laboratory analysis.

    ix. Ease protocol and report preparation through the definition of accepted formats.

    x. Be used as a bid document when soliciting bids for contract execution.

    2.5 Validation Process

    Each part of the validation process should be documented. There should be a written plan for

    performing each validation to specify who is responsible for managing and performing the

    various validation tasks such as production of validation protocols and approvals of validation

    documentation. Validation protocols should be written for each phase of the validation to include

    acceptance criteria. The validation plan and the validation protocols may be combined into a

    single document. The outcome of each phase of validation should be recorded and the overall

    conclusions, with a scientific assessment of any failures should be documented in a validation

    summary report. The validation records and summary report must be reviewed and approved

    before putting the process or system affected into use.

    2.6 Validation Plan

    The plan should first identify the following things:

    What is being validated

    Where the validation will take place

    Why the validation is taking place providing reference to any relevant change control

    records, risk assessments, URS and FDS.

    The validation stages required

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    26

    Validation time-frames

    The plan should also identify the validation team and define responsibilities for :

    Overall management of the validation

    Production of protocols

    Performing the validation and recording the outcome

    Reviewing and approving the protocols and validation records

    Reviewing the validation outcomes and signing off the validation as acceptable.23

    2.7 Typical validation master plan structure

    There is no standard format for master plans. Various authors used different types of plans with

    appropriate adaptations to suit to specific requirements of a particular project. The most

    successfully used plans basic template is given in table below (table 3)24. It can be readily

    modified to different project types and scales. With changes in the facility type, there is a

    corresponding change in the focus of the master plan.

    Table 3 Validation master plan template

    Introduction Introduction to the project scope, location, and timing. Includes

    responsibilities for protocol, SOP, report and other documentation

    preparation and approval. Identifies who is responsible for the

    various activities. A general validation SOP or policy statement may

    be included.

    Plant/Process/Product

    Development

    A concise description of the entire project is provided. It will

    provide information on layout and flow of personnel, materials, and

    components; utility and support systems; description of the

    processes to be performed and products to be made in the facility.

    Major equipment is also described.

    Computerized System

    and Process Control

    Description (If needed)

    Computerized information, laboratory and process control systems

    are described in sufficient detail to delineate the validation

    requirements. This section may be omitted if the level of automation

    is minimal.

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    27

    List of Systems/

    Processes/Products to

    be validated

    Equipment, systems, and products are listed in a matrix format that

    describes the extent of validation required (i.e. IQ, OQ, or PQ) as

    part of the project. Additional breakout of computerized, cleaning

    and sterilization validation requirements can be added.

    General and Specific

    Acceptance Criteria

    Key acceptance criteria (general and specific) for the items listed in

    the prior section are provided. Emphasis should be placed on

    quantitative criteria throughout. To merely state the general

    requirements provides no substantial benefit to either those

    responsible for the validation or for those involved in the design

    process.

    Special Issues (if

    needed)

    Sections can be included describing in greater detail the validation

    requirements of an element of the project where additional

    clarification may be warranted. Typical subjects include automation,

    cleaning, containment, isolation, or lyophilization.

    Protocol and

    Documentation format

    The format to be used for protocols, reports, and operating

    procedures is described. This particularly useful in a new

    organization where such formats have not yet been defined. It can

    also be beneficial when working with an outside contractor to ensure

    that all documentation is in the correct format.

    Required procedures List of SOPs (new or existing) necessary to operate the facility.

    Manpower planning

    and scheduling

    An estimate of the staffing requirements to complete the validation

    effort described in the plan. A preliminary schedule of required

    activities is prepared to help estimate appropriate manning levels.

    2.8 Validation Protocol

    Validation protocol is the step that comes after validation plan. It is an integral element of the

    validation plan. The protocol describes:

    The qualification/validation phase (IQ,OQ, PQ or method process validation)

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    28

    The tests that will be performed

    The test procedures

    The objectives of the validation in terms of acceptance criteria for each test

    Records to be completed.

    What needs to be tested, how many tests to do and the acceptance criteria at each validation

    phase will be specific to each validation and must be founded on the scientific and technical

    basis of the processes and systems involved. It should be possible to establish the specific

    requirements by reference to the relevant risk assessments, URS, FDS, published standards,

    regulations & guidelines.25

    The detailed protocols for performing validations are essential to ensure that the process is

    adequately validated. It should include the following elements:

    Objectives, scope of coverage of the validation study.

    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.

    Critical process parameters and their respective tolerances.

    Process variables and attributes with probable risk and prevention shall be captured.

    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

    results.

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    29

    There should also be a description of the way in which the results will be analyzed. The protocol

    should be approved prior to use. Any changes to a protocol should be approved prior to

    implementation of the change. The validation protocol and report may also include copies of the

    product stability report or a summary of it, validation documentation on cleaning, and analytical

    methods.

    2.9 Validation set up

    Validation set up is very essential to establish the desired attributes. These attributes include

    physical as well as chemical characteristics. In the case of parenterals, these attributes should

    include stability, absence of pyrogens, and freedom from visible particles.

    Acceptance specifications for the product should be established in order to attain uniformity and

    consistently the desired product attributes, and the specifications should be derived from testing

    and challenge of the system on sound statistical basis during the initial development and

    production phases and continuing through subsequent routine production.

    The process and equipment should be selected to achieve the product specification. For e.g.

    design engineers; production and quality assurance people may all be involved. The process

    should be defined with a great deal of specificity and each step of the process should be

    challenged to determine its adequacy. These aspects are important in order to assure products of

    uniform quality, purity and performance.26

    2.10 Relationship between validation and qualification

    Validation and qualification are essential components of the same concept. The term

    qualification is normally used for equipment, utilities and systems, and validation for processes.

    In this sense, qualification is part of validation. Qualification is pre-requisite of validation. There

    are four phases/stages of qualification for process, equipment, facilities or systems:

    Design qualification (DQ);

    Installation qualification (IQ)

    Operational qualification (OQ)

    Performance qualification (PQ)

    Component qualification (CQ), only sometimes stated.

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    30

    All SOPs for operation, maintenance and calibration should be prepared during qualification.

    Training should also be provided to operators and training records be maintained.

    2.10.1 Design Qualification (DQ)

    DQ is defined as Providing documented verification that all key aspects of the design,

    procurement and installation adhere to the approved design intention and that all the

    manufacturers recommendations have been suitably considered.27 It defines the functional and

    operational specifications of the instrument and details the conscious decisions in the selection of

    the supplier.28

    DQ covers all aspects of the design and procurement of facility and equipment. It

    is intended to encompass all those activities that might take place in the design phase, detailed

    and development, including activities associated with procurement of equipment and checkout at

    the suppliers works. It is a verification that the design meets user requirements with a particular

    focus on those requirements that relate to GMP and product quality. The extent of DQ may

    depend on the contract arrangements. Design may be subcontracted to suppliers or

    subcontractors and how this is covered should be defined in the plan. DQ is not a regulatory

    requirement but a smart activity to include in the qualification process. It is essential that aspects

    of design are demonstrated in the qualification process as the existing regulations require that

    facility and equipment are of suitable design and appropriate to purpose.

    DQ should also provide documented evidence that the design specifications were met. Any

    validation should start with setting and documenting the specifications for user requirements,

    instrument functions and performance. The specifications of the instruments design should be

    compared with the user requirement specifications. It is a simple rule of thumb: without

    specifications there is no validation. DQ is the most important step in the validation process.

    Errors made in this phase can have a tremendous impact on the workload during later phases.

    Steps for design qualification: The recommended steps that should be considered for inclusion in

    a design qualification are listed below:

    Description of the analysis problem.

    Selection of the analysis technique.

    Description of the intended use of the equipment.

    Preliminary selection of functional and performance or operational specifications (technical,

    environmental, safety).

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    31

    Preliminary selection of the supplier.

    Instrument tests (if the technique is new).

    Final selection of the equipment.

    Final selection of the supplier.

    Development and documentation of the final functional and operational specifications.

    Role of vendor for design qualification

    Although the user of a system has ultimate responsibility for validation, the vendor also plays a

    major role. The validation covers the complete life of a product, starting with the design and

    development. For commercial off the shelf systems, the user has hardly any influence on how the

    software is being developed and validated, but he can check through documentation to see if the

    vendor followed in acknowledged quality process.

    Tasks of the vendor: The vendor should

    Develop and validate software following documented procedures.

    Test the system and document test cases, acceptance criteria and test results.

    Retain the tests protocols and source code for review at the vendors site.

    Provide procedures for IQ and OQ/PV.

    Implement a customer feedback, change control and response system.

    Provide fast telephone, e-mail and/ or on-site support.

    Qualification of the vendor

    As a part of the qualification process, the vendor should be qualified. The question is, how

    should this be done? Is an established and documented quality system enough, for e.g. ISO

    9001? Should there be a direct audit? Is there another alternative between these two extremes?

    There may be situations where a vendor audit is recommended: for e.g. when computer systems

    are being developed for a specific user. However, this is rarely the case for analytical equipment.

    Typically, off-the-shelf systems are purchased from a vendor with little or no customization for

    specific users.

    The exact procedure to qualify a vendor depends very much on the individual situation, for e.g.

    is the system in mind employing mature or new technology? Is the specific system in widespread

    use either within your own laboratory or your company, or are there references in the same

    industry? Does the system include complex computer hardware and software?

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    32

    2.10.2 Installation Qualification (IQ)

    It can be defined as process of obtaining and documenting evidence that equipment has been

    provided and installed in accordance with the specification. IQ establishes that the instrument is

    received as designed and specified, that it is properly installed in the selected environment, and

    that this environment is suitable for the operation and use of the instrument. This involves

    verification of good engineering practice in installation of equipment, and should consider

    electrical safety, safety issues, location siting, and maintenance/calibration schedules and should

    confirm that the installation has been carried out as specified with the appropriate supporting

    documentation. This activity can be delegated to the supplier, provided that the content of the IQ

    document is approved in advance by the laboratory.

    IQ should provide documented evidence that the installation was complete and satisfactory. It

    should also clearly define those areas and items of equipment systems that are to be qualified.

    The purpose specifications, drawings, manuals, spare parts lists and vendor details should be

    verified during installation qualification. Also, control and measuring devices be calibrated.

    Steps for IQ: Steps for IQ include activities prior and during installation of the equipment. The

    recommended steps are as follows:

    a) Before installation

    Obtain manufacturers recommendations for installation site requirements.

    Check the site for the fulfillment of the manufacturers recommendations (utilities such

    as electricity, water and gases and environmental conditions such as humidity,

    temperature and dust).

    Allow sufficient shelf space for the equipment, SOPs, operating manuals and software.

    b) During installation

    Compare equipment, as received, with purchase order (including software, accessories,

    spare parts)

    Check documentation for completeness (operating manuals, maintenance instructions,

    and standard operating procedures or testing, safety and validation certificates).

    Check equipment for any damage.

    Install hardware (computer, equipment, fittings and tubings for fluid and gas connections

    columns in HPLC and GC, power cables, data flow and instrument control cables).

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    33

    Switch on the instruments and ensure that all modules power up and perform an

    electronic self-test.

    List equipment manuals and SOPs.

    Prepare an installation report.

    2.10.3 Operational Qualification (OQ)

    It can be defined as process of obtaining and documenting evidence that installed equipment

    operates within predetermined limits when used in accordance with its operational procedures.

    OQ is the process of demonstrating that an instrument will function according to its operational

    specification in the selected environment. It should provide documented evidence that utilities,

    systems or equipment and all its components operate in accordance with operational

    specifications. Tests should be designed to demonstrate satisfactory operation over the normal

    operating range as well as at the limits of its operating conditions (including worst case

    conditions). Operation controls, alarms, switches, displays and other operational components

    should be tested and measurements made in accordance with a statistical approach should be

    fully described.29

    It should prove that the instrument is suitable for its intended use. It is not

    required to prove that the instrument meets the manufacturers performance specifications.

    Frequently, people misunderstand and prefer to use the manufacturers specifications because

    usually these are readily available.

    Moreover, this is the verification of process, equipment and facilities over its operating range and

    is assessed against the specifications as defined in the URS. During this stage, a range of tests

    will be carried out to demonstrate the integrity and functionality of the system, including the

    ability to operate under worst case conditions. Confirmation that all calibration, operating and

    cleaning processes have been defined and tested will be required. Definition of the required

    programme of planned preventative maintenance (PPM) should be considered. It can be carried

    out by the supplier and/or by laboratory, or a combination of both. In any case, this must be

    performed using and agreed OQ protocol.

    Steps for OQ

    Define intended functions to be tested.

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    34

    Define test cases and acceptance criteria. For an HPLC system such tests include precision of

    retention times and peak areas, wavelength accuracy of UV detectors, gradient accuracy and

    precision, system carry over, baseline noise and detector linearity.

    Perform tests and compare the results with the acceptance criteria.

    2.10.4 Performance Qualification (PQ)

    It is defined as process of obtaining and documenting evidence that the equipment, as installed

    and operated in accordance with operational procedures, consistently performs in accordance

    with predetermined criteria and thereby yields product meeting its specifications. Successful

    completion of IQ and OQ is followed by PQ. It can also be defined as documented verification

    that all aspects of a facility, utility or equipment perform as intended in meeting predetermined

    acceptance criteria. This is performed to demonstrate that the process, equipment or facility

    performs as required under routine operational conditions and as defined in the URS. This is

    sometimes referred to as Process validation and is the stage of the exercise when the equipment

    or process is assessed in its practical application, with operational outputs/product being assessed

    for acceptability.30

    It should provide documented evidence that utilities, systems or equipment

    and all its components can consistently perform in accordance with the specifications under

    routine use.

    This is generally applicable to those systems that require extended testing over a period of time

    such as water systems, heating, and ventilation systems such as those applicable to clean rooms

    and the actual performance of the clean room to meet the defined standards of operation over

    periods of time. Some organizations may include PQ in the OQ.

    PQ should include following, however, it is not exclusive.

    Tests using production materials, substitutes or simulated product.

    Tests to include condition(s) with upper and lower limits. It will be useful to briefly discuss

    process capability design and testing and process qualification.

    Check actual product and process parameters and procedures established in OQ.

    Test acceptability of the product.

    Check process repeatability, and long term process stability.

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    35

    2.10.5 Component Qualification (CQ)

    It is relatively new term developed in 2005. This refers to the manufacturing of auxiliary

    components to ensure that they are manufactured to the correct design criteria. This could

    involve packaging components such as folding cartons, shipping cases, labels or even phase

    change material. All of these components must have some type of random inspection to ensure

    that the third party manufacturers process is consistently producing components that are used in

    the world of GMP at drug or biologic manufacturer.31

    2.10.6 Requalification

    Requalification should be done in accordance with a defined schedule. The frequency of

    requalification may be determined on the basis of factors such as the analysis of results relating

    to calibration, verification and maintenance. There should be periodic requalification, as well as

    requalification after changes (such as changes to utilities, systems, equipment; maintenance

    work; and movement). It should be considered as part of the change control procedure.

    2.10.7 Revalidation

    Revalidation can be defined as repeating the original validation effort or any part of it, which

    includes investigative review of existing data. It is essential to maintain the validated status of

    the plant, equipment, manufacturing processes and computer systems. Processes and procedures

    should be revalidated to ensure that they remain capable of achieving the intended results. There

    should be periodic revalidation, as well as revalidation after changes. It should be done in

    accordance with a defined schedule. The frequency and extent of revalidation should be

    determined using a risk based approach together with a review of historical data.

    Periodic revalidation should be performed to assess process changes that may occur gradually

    over a period of time, or because of wear of equipment. The following should be considered

    when periodic revalidation is performed:

    Master formulae and specifications

    SOPs

    Records (e.g. of calibration, maintenance and cleaning)

    Analytical methods

  • A REVIEW ON PHARMACEUTICAL VALIDATION

    36

    2.10.8 Revalidation after change

    Revalidation should be performed following a change that could have an effect on the process,

    procedure, quality of the product and/or the product characteristics. Revalidation should be

    considered as part of the change control procedure. The extent of revalidation will depend on the

    nature and significance of the changes. Changes should not adversely affect product quality or

    process characteristics. The changes requiring revalidation should be defined in the validation

    plan, and it may include following:

    Changes in starting materials (including physical properties, such as density, viscosity or

    particle size distribution that may affect the process or product)

    Change of starting material manufacturer

    Transfer of processes to a different site (including change of facilities and installations which

    influence the process)

    Changes of primary packaging material (e.g. substituting plastic for glass)

    Changes in the manufacturing process (e.g. mixing times or drying temperatures)

    Changes in the equipment (e.g. addition of automatic detection systems, installation of new