B Pharm Sem 3
B Pharm Sem 3
B Pharm Sem 3
Topic
Quality Assurance and Quality Management Concepts- Definition and concept
tools .
ISO 9000 & ISO14000- Overview, Benefits, Elements, steps for registration.
QUALITYASSURANCE
According to WHO, quality assurance is a wide- ranging concept covering all matters that
individually or collectively influence the quality of a product. With regard to pharmaceuticals,
quality assurance can be divided into major areas: development, quality control, production,
distribution, and inspections.
ISO 9000 defines as "part of quality management focused on providing confidence that quality
requirements will be fulfilled"
QualityControl
ISO 9000 defines quality control as "A part of quality management focused on fulfilling quality
requirements". It is that part of GMP concerned with sampling, specification & testing,
documentation & release procedures which ensure that the necessary & relevant tests are performed
& the product is released for use only after ascertaining it‟s quality.
Difference between QA and QC
QA aims to prevent defects with a focus on QC aims to identify (and correct) defects
the process used to make the product. It is a in the finishedproduct.
proactive quality process. Quality control, therefore, is areactive
process.
Responsibilities of QA
The QA department is responsible for ensuring that the quality policies adopted by a
company arefollowed.
It helps to identify and prepare the necessary SOPs relative to the control ofquality.
It must determine that the product meets all the applicable specifications and that it was
manufactured according to the internal standards ofGMP.
QA also holds responsible for quality monitoring or auditfunction. QA functions to assess
operations continually and to advise and guide them towards full compliance with
allapplicable internal and externalregulations.
Responsibilities of QC
QC is responsible for the day-to-day control of quality within thecompany.
This department is responsible for analytical testing of incoming raw materials and
inspection of packaging components, includinglabelling.
They conduct in-process testing when required, perform environmental monitoring, and
inspect operations forcompliance.
They also conduct the required tests onfinished dosageform.
QC plays a major role in the selection of qualified vendors from whom raw materials are
results, microbiological tests and release procedure for passed raw materials and components.
Finished Goods- Laboratory Testing and Documentation
This SOP describes the procedure for sampling, location, pre-testing, testing and
documentation of all finished products subject to test, reagents and standards to be used for
analysis, management of out of specification results, microbiological tests and release
procedure for passed finishedgoods.
Total Quality Control (TQC)
The concept of total quality control refers to the process of striving to produce a perfect product
by a series of measures requiring an organized effort at every stage inproduction.
Although the responsibility for assuring product quality belongs principally to QA personnel,
itinvolves many departments and disciplines within acompany.
To be effective, it must be supported by team effort.
Quality must be built into a drug product during product and process, and it is influenced by the
physical plant design, space, ventilation,cleanliness and sanitation during routineproduction.
In products and process designing, itconsiders many parameters like:
Materials
In-process and productcontrol
Specification and tests for active ingredients, excipients
Specific stability procedures of theproduct
Freedom from microbial contamination and properstorage
Containers, packaging andlabeling
Product protection from moisture, light, volatility, and drug/package interaction
pharmaceutical industries. From this concept Total Quality Management (TQM) was established.
The aim of TQM is prevention of defects rather than detection of defects. So TQM is very
important for pharmaceutical industries to produce the better product and ensure the maximum
safety of healthcare system and also protect waste of money for both government &
individualconsumers.
Total Quality Management consists of organization-wide efforts to install and make permanent a
climate in which an organization continuously improves itsability to deliver high-quality products
and services to customers. While there is no widely agreed-upon approach, TQM efforts typically
draw heavily on the previously developed tools and techniques of qualitycontrol.
The production of quality pharmaceuticals products requires embracing the principles of TQM.
Additionally, TQM will serve to improve productivity and customersatisfaction.
The concept of TQM requires the total commitment of senior level managementand supervision
of all departments, operators, suppliers, andcostumers.
It continually strives for process improvement that begins with product development and only
concludes when feedback and follow-up have been completed.
Activities in TQM
TQM is the foundation for activities, which include:
Commitment by senior management and allemployees
Meeting customerrequirements
Reducing development cycletimes
Just in time/demand flowmanufacturing
Improvementteams
Reducing product and servicecosts
Systems to facilitateimprovement
Line managementownership
Employee involvement andempowerment
Recognition andcelebration
Challenging quantified goals andbenchmarking
Focus on processes / improvementplans
Specific incorporation in strategicplanning
Functions of TQM
Product quality criteria are established, and detailed specifications are written. Meticulous,
written procedures must be prepared for production and control. Raw material must be
characterized and then purchased from reputable, approvedsuppliers.
Facilities must be designed, constructed, and controlled to provide the proper stable
environment for protecting the integrity of products. Equipment must be selected that is
efficient and can be cleaned readily and sanitized.
Personnel must be trained properly. The directions they use must be in writing, approved by
responsibleindividuals.
Distribution departments are responsible for controlling the shipping and handling of products,
using inventory-control systems.
The marketing department mustbe sensitive to the costumers‟ needs and be responsive to
complaints.
Advantages of TQM
Improves reputation- faults and problems arespotted and sortedquicker.
Higher employee morale- workers motivated by extra responsibility, team work and
involvement indecisions ofTQM
Lower cost- decrease waste as fewer defective products and no need forseparate.
Quality controlinspector
Disadvantages of TQM
Initial introductioncost.
Benefits may not be seen for several years.
Workers may be resistant tochange
PHILOSOPHY OF TQM
The Philosophy of TQM was born out of the concepts developed by namely four great gurus of
Quality management.
W. Edwards Deming
Joseph M Juran
Armand V Feigenbaum
Philip Crosby
W. Edwards Deming
Deming‟s argument was that quality that is achieved though a reduction in statistical
variation improves competitive position as well as productivity.
He defined Quality as being the direct result of quality of design, quality of conformance
and the quality of the sales and service function.
A great believer in measuring quality by direct statistical measurement against
specification, the goal of quality improvement is to reduce variation.
He developed a set of 14 points for management that express these issues. His beliefs were
that quality management and improvement were the responsibility of all employees in a company.
Deming also believed that managers must change and to develop partnerships with those at
the operating level of the business, one of the key elements in the Total Quality Management
Philosophy.
Joseph Juran
Juran was probably the greatest contributor to the Total Quality Management Philosophy.He
developed his ten-point plan which is the backbone of TQM implementation nowadays.
The Juran Method:
1. Build awareness of the need and opportunity for improvement
2. Set goals for improvement
3. Organize to reach the goals
4. Provide training
5. Carry out projects to solve problems
6. Report progress
7. Give recognition
8. Communicate results
9. Keep the score
10. Maintain momentum by making annual improvement part of the regular system and
processes of the company.
Juran defined Quality as being “Fitness for Use” and really emphasized the cost of quality.
He believed that it was important to take management structure as a starting point and to build the
quality improvement programme from that baseline.
Armand Feigenbaum
Feigenbaum was the originator of the term “Total Quality Control”. He believed that significant
quality improvement could only be achieved by the participation of everyone in the organization.
Fire-fighting quality management should be replaced with clear, customer-oriented quality
management which the employees understand and can commit themselves to.
Feigenbaum believed that the goal of Quality improvement was to reduce the total cost of quality
to as low a percentage as possible.
Philip Crosby
Philip Crosby‟s argument is that higher quality will ultimately reduce costs. He defined Quality as
being the “Conformance to Requirements”.
He developed a programme with 14 steps that has the focus of changing an organization using
action plans for their implementation.
His absolute beliefs were that
1. Quality means conformance and not elegance
2. It is always cheaper to do a job right first time round
3. The only performance indicator is the cost of quality
4. The only performance standard is Zero Defects.
ICH GUIDELINES
ICH Guidelines were created by The International Council for Harmonization of
Technical Requirements for Pharmaceuticals for Human Use (ICH). ICH aims to provide
uniform standards for technical requirements for pharmaceuticals for human use. They are
developed by regulatory and pharma industry authorities.
Mission
To make recommendations towards achieving greater harmonization in the interpretation and
application of technical guidelines and requirements for pharmaceutical product registration,
thereby reducing duplicating of testing carries out during the research and development of new
Human Medicines.
Need to Harmonize
Realization was driven by tragedies, such as that with thalidomide in Europe in the
1960s.
The 1960s and 1970s saw a rapid increase in laws, regulations and guidelines for
reporting and evaluating the data on Quality, Safety and Efficacy of new Medicinal
products.
Divergence in technical requirements from country tocountry
Storage condition
Stability commitment
Evaluation
Statements / labeling
Drug product
The design of the formal stability studies for the drug product should be based on knowledge of
the behavior and properties of the drug substance and from stability studies on the drug substance
and on experience gained from clinical formulation studies. The likely changes on storage and the
rationale for the selection of attributes to be tested in the formal stability studies should be stated.
Includes
Photo stability testing.
Selection of batches.
Container closure system.
Specification.
Testing frequency.
Storage conditions.
Stability commitment.
Evaluation.
Statements/ labeling.
The purpose of this is to outline the changes made in Q1A(R) that result from adoption of ICH
Q1F “Stability Data Package for Registration Applications in Climatic Zones III and IV”.
These changes are:
1. The intermediate storage condition has been changed from 30 °C±2 °C/60% RH±5% RH to
30 °C±2 °C/65% RH±5% RH in the following sections:
Drug Substance - Storage Conditions - General Case
Drug Product - Storage Conditions - General Case
Drug products packaged in semi-permeable containers
2. 30 °C±2 °C/65% RH±5% RH can be a suitable alternative long-term storage condition to
25 °C±2 °C/60% RH±5% in the following sections:
Drug Substance - Storage Conditions - General Case
Drug Product - Storage Conditions - General Case
3. 30 °C±2 °C/35% RH±5% RH has been added as a suitable alternative long-term storage
condition to 25 °C±2 °C/40% RH±5% and the corresponding example for the ratio of water-loss
rates has been included in the following section: Drug products packaged in semi-permeable
containers
4. Mid-stream switch of the intermediate storage condition from 30 °C±2 °C/60% RH±5%
RH to 30 °C±2 °C/65% RH±5% RH can be appropriate provided that the respective storage
conditions and the date of the switch are clearly documented and stated in the registration
application.
It is recommended that registration applications contain data from complete studies at the
intermediate storage condition 30 °C±2 °C/65% RH±5% RH, if applicable, by three years after
the date of publication of this revised guideline in the respective ICH tripartite region.
Q1B Stability testing
Photo stability testing of new drug substances and products
The ICH harmonized tripartite guideline covering the stability testing of new drug substances and
Products (hereafter referred to as the Parent Guideline) notes that light testing should be an integral
part of stress testing. This document is an annex to the Parent Guideline and addresses the
recommendations for photo stability testing.
Drug substances and drug product
Presentation of samples
Analysis of samples
Judgment of results
Immediate (primary) pack is that constituent of the packaging that is in direct contact with the drug
substance or drug product, and includes any appropriate label.
Marketing pack is the combination of immediate pack and other secondary packaging such as a
carton.
Q1C Stability testing for new dosage form
The ICH harmonized tripartite guideline on stability testing of new drug substances and products
was issued on October 27, 1993. This document is an annex to the ICH parent stability guideline
and addresses the recommendations on what should be submitted regarding stability of new dosage
forms by the owner of the original application, after the original submission for new drug
substances and products.
compressing varying amounts of the same granulation, and oral solutions of different strengths
with formulations that differ only in minor excipients (e.g., colorants, flavorings).
With justification, bracketing can be applied to studies with multiple strengths where the relative
amounts of drug substance and excipients change in a formulation. Such justification can include a
demonstration of comparable stability profiles among the different strengths of clinical or
development batches.
In cases where different excipients are used among strengths, bracketing generally should not be
applied.
Matrixing
As defined in the glossary of the parent guideline, matrixing is the design of a stability schedule
such that a selected subset of the total number of possible samples for all factor combinations
would be tested at a specified time point. At a subsequent time point, another subset of samples for
all factor combinations would be tested. The design assumes that the stability of each subset of
samples tested represents the stability of all samples at a given time point. The differences in the
samples for the same drug product should be identified as, for example, covering different batches,
different strengths, different sizes of the same container closure system, and possibly, in some
cases, different container closure systems.
When a secondary packaging system contributes to the stability of the drug product, matrixing can
be performed across the packaging systems.
Each storage condition should be treated separately under its own matrixing design. Matrixing
should not be performed across test attributes. However, alternative matrixing designs for different
test attributes can be applied if justified.
Matrixing designs can be applied to strengths with identical or closely related formulations.
Examples include but are not limited to (1) capsules of different strengths made with different fill
plug sizes from the same powder blend
tablets of different strengths manufactured by compressing varying amounts of the same
granulation, and (3) oral solutions of different strengths with formulations that differ only in minor
excipients (e.g., colorants or flavorings).
Other examples of design factors that can be matrixes include batches made by using the same
process and equipment, and container sizes and/or fills in the same container closure system.
With justification, matrixing designs can be applied, for example, to different strengths where the
relative amounts of drug substance and excipients change or where different excipients are used or
to different container closure systems. Justification should generally be based on supporting data.
For example, to matrix across two different closures or container closure systems, supporting data
could be supplied showing relative moisture vapor transmission rates or similar protection against
light. Alternatively, supporting data could be supplied to show that the drug product is not affected
by oxygen, moisture, or light
Q1E Evaluation of stability data
This guideline is intended to provide recommendations on how to use stability data generated in
accordance with the principles detailed in the ICH guideline “Q1A(R) Stability Testing of New
Drug Substances and Products” (hereafter referred to as the parent guideline) to propose a retest
period or shelf life in a registration application. This guideline describes when and how
extrapolation can be considered when proposing a retest period for a drug substance or a shelf life
for a drug product that extends beyond the period covered by “available data from the stability
study under the long-term storage condition” (hereafter referred to as long-term data).
This guideline addresses the evaluation of stability data that should be submitted in registration
applications for new molecular entities and associated drug products. The guideline provides
recommendations on establishing retest periods and shelf lives for drug substances and drug
products intended for storage at or below “room temperature”*. It covers stability studies using
single- or multi-factor designs and full or reduced designs.
*Note: The term “room temperature” refers to the general customary environment and should not
be inferred to be the storage statement for labeling.
ICH Q6A and Q6B should be consulted for recommendations on the setting and justification of
acceptance criteria and ICH Q1D should be referenced for recommendations on the use of full-
versus reduced-design studies.
The design and execution of formal stability studies should follow the principles outlined in the
parent guideline. The purpose of a stability study is to establish, based on testing a minimum of
three batches of the drug substance or product, a retest period or shelf life and label storage
instructions applicable to all future batches manufactured and packaged under similar
circumstances. The degree of variability of individual batches affects the confidence that a future
production batch will remain within acceptance criteria throughout its retest period or shelf life
Assay procedures are intended to measure the analyte present in a given sample. In the context
of this document, the assay represents a quantitative measurement of the major component(s)
in the drug substance. For the drug product, similar validation characteristics also apply when
assaying for the active or other selected component(s). The same validation characteristics
may also apply to assays associated with other analytical procedures (e.g., dissolution).
The objective of the analytical procedure should be clearly understood since this will govern the
validation characteristics which need to be evaluated. Typical validation characteristics which
should be considered are listed below:
Accuracy
Precision
Repeatability
Intermediate Precision
Specificity
Detection Limit
Quantization Limit
Linearity
Range
Each of these validation characteristics is defined in the attached Glossary. The table lists those
validation characteristics regarded as the most important for the validation of different types of
analytical procedures. This list should be considered typical for the analytical procedures cited but
occasional exceptions should be dealt with on a case-by-case basis. It should be noted that
robustness is not listed in the table but should be considered at an appropriate stage in the
development of the analytical procedure.
Furthermore revalidation may be necessary in the following circumstances:
Changes in the synthesis of the drug substance;
Changes in the composition of the finished product;
Changes in the analytical procedure.
The degree of revalidation required depends on the nature of the changes. Certain other changes
may require validation as well
Organic impurities can arise during the manufacturing process and/or storage of the new drug
substance. They can be identified or unidentified, volatile or non-volatile, and include:
Starting materials
By-products
Intermediates
Degradation products
Reagents, ligands and catalysts
Inorganic impurities can result from the manufacturing process. They are normally known and
identified and include:
Reagents, ligands and catalysts
Solvents are inorganic or organic liquids used as vehicles for the preparation of solutions or
suspensions in the synthesis of a new drug substance. Since these are generally of known toxicity,
the selection of appropriate controls is easily accomplished (see ICH Guideline Q3C on Residual
Solvents).
Excluded from this document are: (1) extraneous contaminants that should not occur in new drug
substances and are more appropriately addressed as Good Manufacturing Practice (GMP) issues,
(2) polymorphic forms, and (3) enantiomeric impurities.
Q3B (R2) Impurities in new drug products
This document provides guidance for registration applications on the content and qualification of
impurities in new drug products produced from chemically synthesized new drug substances not
previously registered in a region or member state.
This guideline addresses only those impurities in new drug products classified as degradation
products of the drug substance or reaction products of the drug substance with an excipient and/or
immediate container closure system (collectively referred to as “degradation products” in this
guideline). Generally, impurities present in the new drug substance need not be monitored or
specified in the new drug product unless they are also degradation products.
Impurities arising from excipients present in the new drug product or extracted or leached from the
container closure system are not covered by this guideline. This guideline also does not apply to
new drug products used during the clinical research stages of development. The following types of
products are not covered in this guideline:
Biological/biotechnological products, peptide, oligonucleotides, radiopharmaceuticals,
fermentation products and semi-synthetic products derived there from, herbal products, and crude
products of animal or plant origin. Also excluded from this document are: (1) extraneous
contaminants that should not occur in new drug products and are more appropriately addressed as
good manufacturing practice (GMP) issues, (2) polymorphic forms, and (3) enantiomeric
impurities.
process for the drug substance or drug product. Therefore, this guideline is intended to assist in the
collection of relevant technical information which serves as evidence that the manufacturing
process changes will not have an adverse impact on the quality, safety and efficacy of the drug
product. The document does not prescribe any particular analytical, nonclinical or clinical strategy.
The main emphasis of the document is on quality aspects
Q6A Specifications: test procedures and acceptance criteria, for new drug substances and
new drug products
This guideline is intended to assist to the extent possible, in the establishment of a single set of
global specifications for new drug substances and new drug products. It provides guidance on the
setting and justification of acceptance criteria and the selection of test procedures for new drug
substances of synthetic chemical origin, and new drug products produced from them, which have
not been registered previously in the United States, the European Union, or Japan.
Q6B Specifications: Test procedures and acceptance criteria for biotechnological/biological
products
The principles adopted and explained in this document apply to proteins and polypeptides, their
derivatives, and products of which they are components (e.g., conjugates). These proteins and
polypeptides are produced from recombinant or non-recombinant cell-culture expression systems
and can be highly purified and characterized using an appropriate set of analytical procedures.
Q7 Good manufacturing practice guide for active pharmaceutical ingredients
This document (Guide) is intended to provide guidance regarding good manufacturing practice
(GMP) for the manufacturing of active pharmaceutical ingredients (APIs) under an appropriate
system for managing quality. It is also intended to help ensure that APIs meet the requirements for
quality and purity that they purport or are represented to possess.
In this Guide “manufacturing” is defined to include all operations of receipt of materials,
production, packaging, repackaging, labeling, relabeling, quality control, release, storage and
distribution of APIs and the related controls. In this Guide the term “should” indicates
recommendations that are expected to apply unless shown to be inapplicable or replaced by an
alternative demonstrated to provide at least an equivalent level of quality assurance. For the
purposes of this Guide, the terms “current good manufacturing practices” and “good
manufacturing practices” are equivalent.
on the principles and concepts described in ICH Guidelines on Pharmaceutical Development (Q8),
Quality Risk Management (Q9) and Pharmaceutical Quality System (Q10) as they pertain to the
development and manufacture of drug substance.
Q12 Technical and regulatory considerations for pharmaceutical product lifecycle
management
This guideline provides a framework to facilitate the management of post-approval CMC changes
in a more predictable and efficient manner. It is also intended to demonstrate how increased
product and process knowledge can contribute to a reduction in the number of regulatory
submissions. Effective implementation of the tools and enablers described in this guideline should
enhance industry‟s ability to manage many CMC changes effectively under the firm‟s
Pharmaceutical Quality System (PQS) with less need for extensive regulatory oversight prior to
implementation.
Q13 Continuous manufacturing of drug substances and drug products
The new ICH guideline will establish harmonized scientific and technical requirements needed to
fulfill regulatory expectations for the implementation and assessment of CM to improve access to
medicines. An ICH guideline would facilitate international harmonization and could reduce
barriers to the adoption of CM technology.
Q14 Analytical procedure development
The new guideline is proposed to harmonize the scientific approaches of Analytical Procedure
Development, and to provide the principles relating to the description of Analytical Procedure
Development process. This new guideline is intended to improve regulatory communication
between industry and regulators and facilitate more efficient, sound scientific and risk-based
approval as well as post-approval change management of analytical procedures
Define a target product quality profile that will be used by formulators and process
engineers as a quantitative surrogate for aspects of clinical safety and efficacy during
product development
Gather relevant prior knowledge about the drug substance, potential excipients and process
operations into a knowledge space. Use risk assessment to prioritize knowledge gaps for
further investigation
Design a formulation and identify the critical material (quality) attributes of the final
product that must be controlled to meet the target product quality profile.
Design a manufacturing process to produce a final product having these critical material
attributes.
Identify the critical process parameters and input (raw) material attributes that must be
controlled to achieve these critical material attributes of the final product. Use risk
assessment to prioritize process parameters and material attributes for experimental
verification. Combine prior knowledge with experiments to establish a design space or
other representation of process understanding.
Establish a control strategy for the entire process that may include input material controls,
process controls and monitors, design spaces around individual or multiple unit operations,
and/or final product tests. The control strategy should encompass expected changes in scale
and can be guided by a risk assessment.
Continually monitor and update the process to assure consistent quality.
Design of experiments (DOE), risk assessment, and process analytical technology (PAT)
are tools that may be used in the QbD process when appropriate
Traditional approach & Enhanced QbD approach
Aspects Current QbD
Pharmaceutical Empirical, Random, Focus on Systematic, Multivariate experiments, Focus
Development optimization on control strategy and robustness
Manufacturing Adjustable within design space, managed by
Fixed
Process company‟s quality systems
PAT utilized, Process operations tracked and
Process Control Some in-process testing
trended
ISO 9000:2000
ISO 9000:2000 refers to the ISO 9000 update released in the year 2000.
The ISO 9000:2000 revision had five goals:
1. Meet stakeholder needs
2. Be usable by all sizes of organizations
3. Be usable by all sectors
4. Be simple and clearly understood
5. Connect quality management system to business processes
ISO 9000:2000 was again updated in 2008 and 2015. ISO 9000:2015 is the most current version.
ISO 9000:2015 principles of Quality Management
The ISO 9000:2015 and ISO 9001:2015 standards are based on seven quality management
principles that senior management can apply to promote organizational improvement.
2. Leadership
Establish a vision and direction for the organization
Set challenging goals
Model organizational values
Establish trust
Equip and empower employees
Recognize employee contributions
Learn more about leadership
3. Engagement of people
Ensure that people‟s abilities are used and valued
Make people accountable
Enable participation in continual improvement
Evaluate individual performance
Enable learning and knowledge sharing
Enable open discussion of problems and constraints
Learn more about employee involvement
4. Process approach
Manage activities as processes
Measure the capability of activities
Identify linkages between activities
Prioritize improvement opportunities
Deploy resources effectively
Learn more about a process view of work and see process analysis tools
5. Improvement
Improve organizational performance and capabilities
Align improvement activities
Empower people to make improvements
Measure improvement consistently
Celebrate improvements
Learn more about approaches to continual improvement
family of standards in 1996. In 2004, ISO 14001 underwent to revision and the current revision of
ISO 14001 was published in September 2015.
Series of ISO 14000
Following are the aspects of environmental management addressed by the ISO series:
Environmental Management Systems (EMS)
Environmental Auditing & Related Investigations (EA&RI)
Environmental Labels and Declarations (EL)
Environmental Performance Evaluation (EPE)
Life Cycle Assessment (LCA)
Terms and Definitions (T&D)
What are the compliances to an ISO 14000 EMS?
Following are the compliances for the ISO 14000 EMS:
Assurance to the customers of your commitment to demonstrable environment
management
Public relations must be excellent
Investor criteria must be satisfied and improve access to capital
Insurance must be obtained at a reasonable cost
Image enhancement and market share
Registration requirements must be met with the clients
Cost control must be improved by identifying and eliminating waste and inefficiency
Lessen the incidents end up in liability
Reduction in the consumption of materials and energy
Facilitates the attainment of permits and authorizations
Decrease the cost of Complying with environmental regulations
Relations between industry and government improves
Registration of ISO 14000
The registration of ISO 14000 is a formal recognition of an organization‟s ability to conform to the
requirements of an EMS. Any organization can simply declare that their EMS meets the
requirements of ISO 14001 ( self- declaration). In addition to this, many organizations choose to
have their EMS registered and it happens usually to provide greater assurance to clients and the
public or because regulators and clients require it.
What are the basic Principles behind the ISO 14000 series?
Following are the key principles of the ISO 14000 standards:
1. Result in better environmental management
2. Encompass environmental management system and the aspects of environmental products
3. Applicable in all countries.
4. Promote the broader interests of the public as well as users of these standards.
5. Cost-effective as well as non-perspective and flexible so they are able to meet the differing
needs of organizations of any type or size, worldwide
6. Flexibility to be suitable for internal and external verification
7. Scientifically based
8. Last but not least, Practical, useful and usable.
What are the benefits of getting ISO 14000 Certified?
Following are the benefits of getting the ISO 14000 certification:
1. It identifies and controls the environmental impact of its activities, product or services.
2. Continuously improve its environmental performance
3. Helps in implementing a systematic approach to setting environmental objectives to
achieving these and to demonstrating that they have been achieved.
4. Ensuring legal compliance.
Apart from this, there are several other benefits also which is categorized under Internal benefits
and External benefits:
Internal Benefits:
Assurance to the management: It is in the control of the organizational procedures and
activities having an impact on the environment.
Assure employees: It assures the employees that they are working for an environmentally
responsible organization.
External Benefits:
It provides assurance on environmental issues to the external stakeholders such as
customers, community and regulatory agencies.
Comply with the regulations of the environment.
Claims and communication: It supports the organization‟s claims and communication
about its own environmental policies, plans, and actions by gaining its EMS certificate.
3. Train a person on Quality Management System and Internal Audit (4-day residential training
courses conducted by NABL. Contact NABL Secretariat for details).
4. Prepare QUALITY MANUAL as per ISO 15189 standards.
5. Prepare Standard Operating Procedure for each investigation carried out in the laboratory.
6. Ensure effective environmental conditions (temperature, humidity, storage placement, etc.).
7. Ensure calibration of instruments / equipment. Only NABL ACCREDITED CALIBRATION
LABORATORIES are authorized to provide calibration. NABL website gives the names of
NABL accredited calibration laboratories in the various fields of Accreditation.
8. Impart training on the key elements of documentation, such as document format, authorization
of document, issue and withdrawal procedures, document review and change, etc. Each
document should have ID No., name of controlling authority, period of retention, etc.
9. Ascertain the status of the existing quality system and technical competence with regard to
NABL standards and address the question “Is the system documented and effective OR does it
need modification?”
10. Remember Quality Manual is a policy document, which has to be supplemented by a set of
other next level documents. Therefore ensure that these documents are well prepared.
11. Ensure proper implementation of all aspects that have been documented in the Quality Manual
and other documents.
12. Incorporate Internal Quality Control (IQC) practice while patients‟ samples are analysed.
13. Document IQC data as well as uncertainty of measurements. Maintain Levy Jennings charts.
14. Participate in External Quality Assessment Schemes (EQAS). If this is not available for certain
analytes, participate in inter-laboratory comparison through exchange of samples with NABL
accredited laboratories.
15. Document corrective actions on IQC / EQA outliers.
16. Conduct Internal Audit and Management Review.
17. Apply to NABL along with appropriate fee.
Accreditation Process
An applicant laboratory is expected to submit to NABL 5 copies of the application and 5 copies of
Quality Manual. The Quality Manual will be forwarded by NABL to a Lead Assessor to judge the
adequacy of the Quality Manual as to whether it is in compliance with ISO 15189 standards.
Thereafter the Lead Assessor will conduct a PreAssessment of the laboratory for one day. Based
on the Pre-Assessment report the laboratory may have to take certain corrective actions, so as to be
fully prepared for the final assessment. It is essential for the applicant as well as accredited
laboratories to satisfactorily participatein Proficiency testing/ Interlaboratory comparisons/External
quality assessment programme as Asia Pacific Laboratory Accreditation Cooperation (APLAC)
Mutual Recognition Arrangement calls for mandatory participation in such programmes. Finally
when the laboratory is ready, the Lead Assessor and a team of technical assessors will conduct the
final assessment. The number of technical assessors will depend on the number of disciplines
applied for. The accreditation process involves a thorough assessment of all the elements of the
laboratory that contribute to the production of accurate and reliable test data. These elements
include staffing, training, supervision, quality control, equipment, recording and reporting of test
results and the environment in which the laboratory operates. The laboratory may have to take
certain corrective actions, after the final assessment. After satisfactory corrective actions are taken
by the laboratory (within a period of 3 months), the Accreditation Committee will examine the
report and if satisfied recommend accreditation.
The time required for the process of accreditation will depend upon the preparedness of the
laboratory and its response to the non - conformances raised during the pre-assessment and final
assessment. The total duration ranges between 6 and 8 months.