PHARMACEUTICAL QUALITY MANAGEMENT SYSTEM: A REVIEW

Similar documents
ICH Q8, 9 & 10 and the Impact on the QP

Importance of ICH Guidance in Fulfilling Process Validation Requirements

Challenges of Implementation of ICH Q 8

Implementing Quality Systems

Quality Risk Management

2008 Course Programs Schedule

Quality assurance in the supply chain for pharmaceuticals from the WHO perspective

Value Paper. Are you PAT and QbD Ready? Get up to speed

Impact of ICH Q9 and the application of Risk Management

PRIMATECH WHITE PAPER COMPARISON OF FIRST AND SECOND EDITIONS OF HAZOP APPLICATION GUIDE, IEC 61882: A PROCESS SAFETY PERSPECTIVE

the SPD company Dr Clive Simon, Principal, The SPD Company.

ICH Q12 (Pharmaceutical Product Lifecycle Management): PMDA Perspective

CDER s Office of Pharmaceutical Quality (OPQ): Delivering on the 21 st Century Quality Goals

Claudio Pincus, President, The Quantic Group R. Owen Richards, President, Quantic Regulatory Services Daniel Pincus, Consultant, The Quantic Group

Claus Mortensen, Medicines Inspector. Danish Medicines Agency. Member of the EMEA PAT team

NZFSA Policy on Food Safety Equivalence:

Q8 and Q8 annex An industry Perspective

PDA 71 Years of Connecting People, Science and Regulation

Applied Safety Science and Engineering Techniques (ASSET TM )

Quality by Design. Innovate Design Development Create value. Correct definition of QbD and its relation to product and process development

HTA Position Paper. The International Network of Agencies for Health Technology Assessment (INAHTA) defines HTA as:

Extract of Advance copy of the Report of the International Conference on Chemicals Management on the work of its second session

Quality by Design and OINDP. Today s Presentation

EU GMP Evolution or Revolution Scope and drivers for EU GMP changes. August Gordon Farquharson

OSIsoft. Users Conference 2013

ICH Q10 Pharmaceutical Quality System

Fiscal 2007 Environmental Technology Verification Pilot Program Implementation Guidelines

Future of Pharmaceutical Quality and the Path to Get There

December Eucomed HTA Position Paper UK support from ABHI

Guidance for Industry

Medical Device Risk Management

Guidance for Industry and FDA Staff Use of Symbols on Labels and in Labeling of In Vitro Diagnostic Devices Intended for Professional Use

Swissmedic, Swiss Agency for Therapeutic Products

JUST SCRATCHING THE SERVICE

OMCL Network of the Council of Europe GENERAL DOCUMENT

Intellectual Property, Vaccine Production and Technology Transfer

Innovative Approaches to Pharmaceutical Development and Manufacturing Seminar Series

Global Harmonization Task Force

MAH Responsibilities including the Management of CMOs. QP Forum Workshop. Robert Byrne and Eoin Duff Quality Assurance Alexion Pharmaceuticals

Global GMP Harmonisation A Japanese Perspective

Agency Information Collection Activities; Proposed Collection; Comment Request; Good

Environmental Protection Agency

Issues in Emerging Health Technologies Bulletin Process

WHO Regulatory Systems Strengthening Program

CENTER FOR DEVICES AND RADIOLOGICAL HEALTH. Notice to Industry Letters

This is a preview - click here to buy the full publication

QUALITY: BRACKETING AND MATRIXING DESIGNS FOR STABILITY TESTING OF NEW VETERINARY DRUG SUBSTANCES AND MEDICINAL PRODUCTS

FSMA Update. Jennifer Thomas Interim Director for FSMA Operations Center for Food Safety and Applied Nutrition Food and Drug Administration May 2018

Pharmaceutical Manufacturing and Engineering Catalog Excerpt

ICH Q-IWG Integrated Training Programme

COMMUNICATION FROM THE COMMISSION TO THE EUROPEAN PARLIAMENT. pursuant to Article 294(6) of the Treaty on the Functioning of the European Union

Department of Energy s Legacy Management Program Development

Connecting People, Science and Regulation

peace of mind For from development to commercial supply

Implementation of Directive 2010/63/EU: - the animal welfare perspective

Inter-Association Task Force

Progressive Licensing and the Modernization of the Canadian Regulatory Framework

SAUDI ARABIAN STANDARDS ORGANIZATION (SASO) TECHNICAL DIRECTIVE PART ONE: STANDARDIZATION AND RELATED ACTIVITIES GENERAL VOCABULARY

This document is a preview generated by EVS

CMC Topics and PMDA s activities Yoshihiro Matsuda, Ph.D.

SHTG primary submission process

Current Status and Challenges of Bilateral/Multilateral Meetings

Quality by Design, Clinical Relevance & Lifecycle Considerations

Triennial Review of the Medicines and Healthcare Products Regulatory Agency. Call for Evidence

Global Regulatory Update FDA, USP, EP and ICH Standards for Test Method Validation. Jane Weitzel

Phase 2 Executive Summary: Pre-Project Review of AECL s Advanced CANDU Reactor ACR

E5 Implementation Working Group Questions & Answers (R1) Current version dated June 2, 2006

Office of Pharmaceutical Quality Key Quality Initiatives

PMDA perspective on Quality by Design for pharmaceutical products

MU064: Mechanical Integrity & Reliability in Refineries, Petrochemical & Process Plant

Medical Technology Association of NZ. Proposed European Union/New Zealand Free Trade Agreement. Submission to Ministry of Foreign Affairs & Trade

Terrence Tougas. Dennis Sandell

Technically Unavoidable Particles Profile (TUPPs) Guide

IGDRP Mission, Scope, How it works

Brazilian industry s activities for international convergence. Walker Lahmann Director of External Trade at ABIFINA Executive Director at Eurofarma

Ministry of Justice: Call for Evidence on EU Data Protection Proposals

Systems. Professor Vaughan Pomeroy. The LRET Research Collegium Southampton, 11 July 2 September 2011

February 5, 2010 VIA ELECTRONIC SUBMISSION

Overview on Medicines Regulation: regulatory cooperation and harmonization in focus

FDA s Evolving Approach to Pharmaceutical Quality

This document is a preview generated by EVS

Piloting MDevSPICE - the Medical Device Software Process Assessment Framework

Quality Regulation under Revised Pharmaceutical Affair Law

Functional safety for semiconductor IP

Technology Transfer as the Process of Pharmaceutical Quality System: Modelling Technology Transfer as a Process Strategy

What We Heard Report Inspection Modernization: The Case for Change Consultation from June 1 to July 31, 2012

Office of Pharmaceutical Quality: Why, What, and How?

New Approaches to Safety and Risk Management

EDQM COUNCIL OF EUROPE CONFERENCE CERTIFICATION PROCEDURE : 20 YEARS OF EXPERIENCE March EDQM, Strasbourg, France ABSTRACTS

The International Pharmacopoeia Overview

Technical Documentation - Key pit falls

Masao Mukaidono Emeritus Professor, Meiji University

Essential requirements for a spectrum monitoring system for developing countries

Verification & Validation

Recast de la législation européenne et impact sur l organisation hospitalière

MA/INS/GMP/735037/2014 Annex 1 of the GMP Guidelines on Good Manufacturing Practice - Manufacture of Sterile Medicinal Products

BUREAU OF LAND MANAGEMENT INFORMATION QUALITY GUIDELINES

28 March Report of the Working Group on Pharmaceuticals and Public Health of the High Level Committee on Health.

NIHR / Wellcome Trust King s Clinical Research Facility. Guidance for Investigators

Keynote GMP & Validation from disaster, via overkill, to common sense.

Transcription:

PHARMACEUTICAL QUALITY MANAGEMENT SYSTEM: A REVIEW Available online at www.ijdra.com REVIEW ARTICLE Sharma S*, Bodla R.B., Shweta S, Pathak D.P. Delhi Institute of Pharmaceutical Sciences and Research, New Delhi, India *Corresponding Author s E-mail: sharmashrey7@gmail.com ABSTRACT ICH is International Conference on Harmonization of technical requirements for registration of pharmaceuticals for human use. The goal of ICH is to discuss and establish common guidelines by bringing together three ICH regions i.e. USA, Japan and EU and to make information available on ICH, its activities and guidelines. The guidelines have become more relevant for Generic s Drug Approval. Drug Development and manufactures are more and more global. Harmonization is considered more important than ever, as it facilitates work sharing, efficient use of resources and access to medicines. The objective of harmonization is meant for economical use of human, animal and other resources and the elimination of unnecessary delay in the global development and of availability of new medicines whilst maintaining safeguards on quality, safety and efficacy. The present discussion of the review is on ICH guidelines for quality control focussing mainly on Quality Risk Management (Q9). Quality control is a process that is used to ensure a certain level of quality in a product or service. It might include whatever action a business deems necessary to provide for the control and verification of certain characteristics of a product or service. Most often, it involves thoroughly examining and testing the quality of products or the results of services. The basic goal of this process is to ensure that the products or services that are provided meet specific requirements and characteristics, such as being dependable, satisfactory, safe and fiscally sound. Quality risk management is a systematic process for the assessment, control, communication and review of risks to the quality of the drug product across the product lifecycle. Two primary principles of quality risk management are: The evaluation of the risk to quality should be based on scientific knowledge and Ultimately linked to the protection of the patient; and The level of effort, formality, and documentation of the quality risk management process should be commensurate with the level of risk. Keywords: Quality Risk Management (Q9), ICH, Quality risk management. INTRODUCTION ICH is a joint initiative involving both Regulators and Research-based industry initiatives of the Europe, Japan and US for the scientific and technical discussions of the testing procedures; required to assess and ensure the Safety, Quality and Efficacy of the medicines. ICH Stands for International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for Human use. (1) Aim ICH was established in 1990, as a joint regulatory/ industry project to improve, through harmonization, the efficiency of the process, for developing and registering new medicinal products in Europe, Japan and US. To make medicinal products available to the patients with minimum of a delay. (1) Need of ICH At the time, the Pharma market had become increasingly international and Industry was seeking to market its products internationally. The 3 major Pharma market were USA, Europe and Japan- the Triad. While all three had Drug Regulatory approval systems and that too were based on same principles. This means-the detailed technical requirements in each region differed. (1) For the Industry, this meant: 2014 IJDRA Publishing Group, All rights reserved Page 67

Duplicate test procedures. Duplicate test procedures were time consuming and Expensive. Submitting different and Huge (Lorry-sized) applications due to which a NDA were considered as Nightmare. This led to: 1. Rising costs of health care. 2. Escalation of the cost of Research and Development. Hence, need to meet the public expectation that there should be a minimum of delay in making safe and efficacious new treatments available to patients have arisen. Hence, there was an urgent need to rationalize and harmonize regulation. (1) HISTORY EU took initiative of harmonization of regulatory requirements in 1980. At the same time, bilateral discussions between Europe, Japan and US on possibilities for Harmonization at WHO Conference of Drug Regulatory authorities, in Paris, in 1989, specific plans for action began to materialize. Authorities approached IFPMA (International Federation of Pharmaceutical Manufacturers and Associations) to discuss joint regulatoryindustry initiative on international harmonization and ICH was conceived. The birth of ICH took place at a meeting in April 1990, hosted by the EFPIA (European Federation of Pharmaceutical Industries and Association) in Brussels. (1) ICHs decades As the 1 st decade Significant progress in the development of Tripartite ICH Guidelines on Safety, Quality and Efficacy topics Work has also undertaken on number of important multidisciplinary topics- MedDRA (Medical Dictionary of Regulatory Activities) CTD (Common Technical Document) As the 2 nd decade The development of ICH guidelines continued, but with more attention given to the following need of: Maintain already existing Guidelines as Science and Technology continued to evolve; Expand communication and dissemination of information on ICH Guidelines. As the 3 rd decade ICHs attention is directed towards extending the benefits of harmonization beyond the ICH regions. (1) ORGANIZATION OF ICH Figure 1: Organization of ICH 2014 IJDRA Publishing Group, All rights reserved Page 68

Goals of ICH Figure 2: Goals of ICH Purpose of ICH The basic purpose of ICH are- To monitor, update and increase the international harmonization of Technical Requirements. To ensure Safety, Efficacy and Quality of medicines that must be developed and registered in the most efficient and cost effective manner. Quality Topics To promote and protect public health from an international perspective. To prevent unnecessary duplication of clinical trials in humans. To minimize the use of animal testing without compromising the safety and effectiveness. To improve the efficiency of Global Drug Development. (1) Table 1: Quality Topics Q1 Q2 Q3 Q4 Q5 Q6 Q7 Q8 Q9 Q10 STABILITY ANALYTICAL VALIDATION IMPURITES PHARMACOPOEIAS QUALITY OF BIOTECHNOLOGICAL PRODUCTS SPECIFICATIONS GMPs PHARMA. DEVELPOMENT QRM PHARMA. QUALITY SYSTEM 2014 IJDRA Publishing Group, All rights reserved Page 69

ICH GUIDELINES Q - Quality Topics i.e. those related to Chemical & Pharmaceutical Quality Assurance. S - Safety Topics i.e. those related to In-Vitro & In-vivo pre-clinical studies. E - Efficacy Topics i.e. those related to clinical studies in Human Subject. M - Multidisciplinary Topics i.e. cross-cutting topics which do not fit uniquely into one of the above categories. Figure 3: ICH Guidelines Q9- QUALITY RISK MANAGEMENT (2) Definition Quality: Degree to which a set of inherent properties of a product, system or process fulfills requirements. Risk: Combination of the probability of occurrence of harm and the severity of that harm. MANAGEMENT Systematic process for the assessment, control, communication and review of risks to the quality of the drug (medicinal) product across the product lifecycle. QRM Figure 4: QRM Management PRINCIPLES OF QUALITY RISK MANAGEMENT Two primary principles of quality risk management are: 2014 IJDRA Publishing Group, All rights reserved Page 70 The evaluation of the risk to quality should be based on scientific knowledge and ultimately link to the protection of the patient; and

The level of effort, formality, and documentation of the quality risk management process should be commensurate with the level of risk. SCOPE OF QRM This guidance provides principles and examples of tools for quality risk management that can be applied to different aspects of pharmaceutical PROCESS OF QRM (2) quality. These aspects include development, manufacturing, distribution, inspection, and submission/review processes throughout the lifecycle of drug substances, drug products, biological and biotechnological products (including the use of raw materials, solvents, excipients, packaging and labelling materials in drug products, biological and biotechnological products). Figure 5: Process of QRM RISK MANAGEMENT METHODS AND TOOLS (2) 1. Basic Risk Management Facilitation Methods Some of the simple techniques that are commonly used to structure risk management by organizing data and facilitating decision making are: FMEA provides for an evaluation of potential failure modes for processes and their likely effect on outcomes and/or product performance. (3) Potential Areas of Use FMEA can be used to prioritize risks and monitor the effectiveness of risk control activities. 2. Failure Mode Effects Analysis (FMEA) 2014 IJDRA Publishing Group, All rights reserved Page 71

1. Flowcharts 2. Check sheets 3. Cause and Effect Diagrams( fish bone dia) 4. Process Mapping Figure 6: Risk Management methods and Tools 3. Failure Mode, Effects and Criticality Analysis (FMECA) FMEA might be extended to incorporate an investigation of the degree of severity of the consequences, their respective probabilities of occurrence, and their detectability, thereby becoming a Failure Mode, Effects, and Criticality Analysis (FMECA). (4) Potential Areas of Use FMECA application in the pharmaceutical industry should mostly be utilized for failures and risks associated with manufacturing processes. 4. Fault Tree Analysis (FTA) The FTA tool is an approach that assumes failure of the functionality of a product or process. This tool evaluates system (or subsystem) failures one at a time but can combine multiple causes of failure by identifying causal chains. (5) Potential Areas of Use FTA can be used to investigate complaints or deviations in order to fully understand their root cause and to ensure that intended improvements will fully resolve the issue and not lead to other issues (i.e. solve one problem yet cause a different problem). Fault Tree Analysis is an effective tool for evaluating how multiple factors affect a given issue. 5. Hazard Analysis and Critical Control Points (HACCP) It is a structured approach that applies technical and scientific principles to analyze, evaluate, prevent, and control the risk or adverse consequence(s) of hazard(s) due to the design, development, production, and use of products. (6) 6. Hazard Operability Analysis (HAZOP) HAZOP is based on the assumptions that risk events are caused by deviations from the design or operating intentions. It is a systematic brainstorming technique for identifying hazards. 7. Preliminary Hazard Analysis (PHA) PHA is a tool of analysis based on applying prior experience or knowledge of a hazard or failure to identify future hazards, hazardous situations and events that might cause harm, as well as to estimate their probability of occurrence for a given activity, facility, product, or system. 2014 IJDRA Publishing Group, All rights reserved Page 72

8. Risk Ranking and Filtering Risk ranking and filtering is a tool for comparing and ranking risks. Risk ranking of complex systems typically involves evaluation of multiple diverse quantitative and qualitative factors for each risk. Filters, in the form of weighting factors or cut-offs for risk scores, can be used to scale or fit the risk ranking to management or policy objectives. 9. Supporting Statistical Tools They can enable effective data assessment, aid in determining the significance of the data set(s), and facilitate more reliable decision making. IMPLEMENTATION OF QRM Figure 7: Implementation of QRM HURDLES IN ABSENCE OF QRM Figure 8: Hurdles in absence of QRM 2014 IJDRA Publishing Group, All rights reserved Page 73

The absence of a harmonised risk management approach has the following impacts on the patient, regulator and/or industry: Product may not be available to patients, when needed. May increase the potential for the release of unacceptable product to the market New product introductions to the marketplace may be delayed. Delays may occur during implementation of changes and improvements to processes. Safe and effective drugs may be discarded or recalled from the market. Manufacturers may be reluctant to implement new technologies or continuous improvements to the products or processes. Scarce resources may not be optimally allocated. Lack of appropriate data to evaluate risk most effectively Issues to be Resolved The following issues need to be resolved: Terminology including a definition of quality, risk, risk management, etc Principles for how risk management should be effectively applied and consistently integrated into decisions regarding product quality and impact on the patient. Identifying circumstances when applying risk management principles is not feasible or appropriate. Defining what principles of risk management apply to the industry, to the regulators, and to both, throughout the product lifecycle. How, what and when information is exchanged between and within industry and regulators, in a global context. How to ensure synergy with the Pharmaceutical Development EWG and the resulting guidelines. Defining roles and responsibilities of regulators and industry, including communication responsibilities. BENEFITS How risk can be incorporated into resource allocation decisions. Benefits of the harmonized risk management guideline to all ICH parties and observers: Enhanced patient confidence in decision making on pharmaceutical quality Promotes more effective use of regulatory agency and industry resources Establishes a systematic, well-informed and thorough method of decision making which leads to greater transparency and predictability Increased knowledge of exposure to risk Fosters quality by design, continuous improvement and new technology introduction, which generally leads to enhanced product quality Enhanced empowerment and flexibility in work. POTENTIAL APPLICATIONS FOR QUALITY RISK MANAGEMENT (2) 1. Quality Risk Management as Part of Integrated Quality Management Documentation To review current interpretations and application of regulatory expectations. To determine the desirability of and/or develop the content for SOPs, guidance, etc. Training and education To determine the appropriateness of initial and/or ongoing training sessions based on education, experience, and working habits of staff, as well as on a periodic assessment of previous training (e.g., its effectiveness). To identify the training, experience, qualifications, and physical abilities that allow personnel to perform an operation reliably and with no adverse impact on the quality of the product. 2014 IJDRA Publishing Group, All rights reserved Page 74

Quality defects To provide the basis for identifying, evaluating, and communicating the potential quality impact of a suspected quality defect, complaint, trend, deviation, investigation, out of specification result, etc. To facilitate risk communications and determine appropriate action to address significant product defects, in conjunction with regulatory authorities (e.g., recall). Figure 9: Benefits of the harmonized risk management guideline Auditing/Inspection To define the frequency and scope of audits, both internal and external, taking into account factors such as: Existing legal requirements, Overall compliance status and history of the company or facility. Robustness of a company s quality risk management activities etc. Periodic review To select, evaluate, and interpret trend results of data within the product quality review. To interpret monitoring data (e.g., to support an assessment of the appropriateness of revalidation or changes in sampling). Change management/change control To manage changes based on knowledge and information accumulated in pharmaceutical development and during manufacturing. To evaluate the impact of the changes on the availability of the final product. To evaluate the impact on product quality of changes to the facility, equipment, material, manufacturing process, or technical transfers. To determine appropriate actions preceding the implementation of a change, e.g., additional testing, (re)qualification, (re)validation, or communication with regulators. 2. Quality Risk Management as Part of Regulatory Operations Inspection and assessment activities To assist with resource allocation including, for example, inspection planning and frequency, and inspection and assessment intensity. To evaluate the significance of, for example, quality defects, potential recalls, and inspectional findings. To determine the appropriateness and type of post inspection regulatory follow-up. 2014 IJDRA Publishing Group, All rights reserved Page 75

To evaluate information submitted by industry, including pharmaceutical development information. To evaluate impact of proposed variations or changes. To identify risks that should be communicated between inspectors and assessors to facilitate better understanding of how risks can be or are controlled (e.g., parametric release, Process Analytical Technology (PAT). Figure 10: Quality Risk Management as part of Regulatory Operations 3. Quality Risk Management as Part of Materials Management Assessment and evaluation of suppliers and contract manufacturers To provide a comprehensive evaluation of suppliers and contract manufacturers (e.g., auditing, supplier quality agreements). Starting material To assess differences and possible quality risks associated with variability in starting materials (e.g., age, route of synthesis). Use of materials To determine whether it is appropriate to use material under quarantine (e.g., for further internal processing). To determine appropriateness of reprocessing, reworking, use of returned goods. Storage, logistics and distribution conditions To assess the adequacy of arrangements to ensure maintenance of appropriate storage and transport conditions (e.g., temperature, humidity, container design). To determine the effect on product quality of discrepancies in storage or transport conditions(e.g., cold chain management) in conjunction with other ICH guidance. To maintain infrastructure (e.g., capacity to ensure proper shipping conditions, interim storage, handling of hazardous materials and controlled substances, customs clearance). To provide information for ensuring the availability of pharmaceuticals (e.g., ranking risks to the supply chain). 4. Quality Risk Management as Part of Production Validation 2014 IJDRA Publishing Group, All rights reserved Page 76

To identify the scope and extent of verification, qualification, and validation activities (e.g. analytical methods, processes, equipment, and cleaning methods). To determine the extent for followup activities (e.g., sampling, monitoring, and re-validation). To evaluate the frequency and extent of in-process control testing (e.g., to justify reduced testing under conditions of proven control). To evaluate and justify the use of process analytical technologies (PAT) in conjunction with parametric and real time release. To distinguish between critical and noncritical process steps to facilitate design of a validation study. (7) In-process sampling and testing Production planning To determine appropriate production planning (e.g., dedicated, campaign, and concurrent production process sequences). Figure 11: Managing risk of Drug Product use 5. Quality Risk Management as Part of Laboratory Control and Stability Studies Out of specification results To identify potential root causes and corrective actions during the investigation of out of specification results. (8,9) Retest period/expiration date To evaluate adequacy of storage and testing of intermediates, excipients, and starting materials. 6. Quality Risk Management as Part of Packaging and Labelling Design of packages To design the secondary package for the protection of primary packaged product (e.g., to ensure product authenticity, label legibility). Selection of container closure system To determine the critical parameters of the container closure system. Label controls To design label control procedures based on the potential for mix-ups involving different product labels, 2014 IJDRA Publishing Group, All rights reserved Page 77

including different versions of the same label. CONCLUSION 1. Over all Positive Contribution is towards patient protection Further develops Quality Risk Management awareness that is already part of industry and regulatory culture. 2. Ongoing change in behaviour Identifying risks can be positive A long list of identified risks that are assessed and controlled provides high quality capability Awareness of quality risks Risk-based approach A potential of risks remains - No Zero risk! ACKNOWLEDMENT This is matter of great pride to pursue my Master of Pharmacy from Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR). I would like to express my sincere gratitude to my guide Dr. Ramesh B. Bodla, (Associate Professor), who gave me the appropriate guidance and has shown interest and encouraged me to carry out work on this review article. www.icssc.org/presentations/newdelhi2007/3briefin trototheichguidelinesindia2007.pdf 2. Guidance for Industry-Center for Drug Evaluation and Research. [Internet] Fishers Lane, Rockville: FDA; 2006 June [cited 2014 June 02]. Available from: http://www.fda.gov/cder/guidance/index.htm 3. Stamatis D H. FMEA from Theory to Execution, 2nd ed. ASQ Quality Press; 2003. p.391-400, ISBN 0873895983. 4. Roberts P. FMECA [Internet].UK: FMECA; 2007 Jan [cited 2014 July 2]. Available from: www2.warwick.ac.uk/fec/sci/wmg/ftmsc/modules/mo dulelist/peuss/slides/section_12a_fmeca_notes.pdf. 5. FTA - Fault tree analysis, 2 nd ed. IEC 61025 [Internet]. 2006 [cited 2014 July 7]. Available from: webstore.iec.ch/preview/info_iec61025%7bed2.0%7 Den_d.pdf. 6. Methodology to pharmaceuticals, Annex-7: Application of Hazard Analysis and Critical Control Point (HACCP). WHO Technical Report. S. No. 908; 2003. 7. Safety aspects - Guideline for their inclusion in standards-2014, no. 51-ISO/IEC Guide [Internet]. 2014 [cited 2014 July 27]. Available from: www.iso.org/obp/ui/#iso:std:iso-iec:guide:51:ed- 3:v1:en. 8. Thrussell IR. Quality Risk Management and its application in sterile processing. WHO [Internet]. WHO; 2009 Nov [cited 2014 July 11]. Available from: apps.who.int/prequal/training resources/3-3_risk_ Management.ppt 9. Risk management - Vocabulary - Guidelines for use in standards-2009, no.73-iso/iec Guide. [Internet]. ISO; 2009 [cited 2014 July 27]. Available from: www.iso.org/obp/ui/#iso:std:iso:guide:73:ed-1:v1:en. I would also like to express thanks to Dr. D. P. Pathak (Director, DIPSAR) for granting me the opportunity to undertake this article on ICH : Quality System Management. I would like to express my gratitude and thanks to Shweta Singh for her continuous support and encouragement and for being a helping hand during this article work. CONFLICT OF INTEREST Author declares that there are no conflict of interest. REFERENCES 1. Mario Chen. Brief Introduction to the ICH Guidelines. Family Health International Biostatistics Workshop [Internet].India; 2007 March [cited 2014 July 15]. Available from: 2014 IJDRA Publishing Group, All rights reserved Page 78