MONTE CARLO SIMULATIONS FOR RISK ANALYSIS IN PHARMACEUTICAL PRODUCT DESIGN

Similar documents
Workshop B Control Strategy

Pharmaceutical Quality Systems: US Perspective

PHARMACEUTICAL DEVELOPMENT

Validation and Calibration. Definitions and Terminology

A NUSAGE-PharmEng Pharmaceutical and Biotechnology Training Program

Evaluating Current Practices in Shelf Life Estimation

Quality by Design Approaches to Analytical Methods -- FDA Perspective. Yubing Tang, Ph.D. FDA/CDER/ONDQA AAPS, Washington DC October 25, 2011

Guidance for Industry PAT A Framework for Innovative Pharmaceutical Development, Manufacturing, and Quality Assurance

C 5. chemical development contract research custom synthesis cgmp API manufacturing commercial production. Welcome to

PROPOSAL FOR REVISION OF THE SUPPLEMENTARY GUIDELINES ON GOOD MANUFACTURING PRACTICES: VALIDATION, APPENDIX 7: NON-STERILE PROCESS VALIDATION

Guideline on Process Validation

Guidance for Industry

Guidance for Industry

Journal of Chemical and Pharmaceutical Research

SPECIFICATIONS AND CONTROL TESTS ON THE FINISHED PRODUCT

QUALITY BY DESIGN (QBD) : A COMPLETE REVIEW

ICH Topic Q 1 A Stability Testing Guidelines: Stability Testing of New Drug Substances and Products

Guidance for Industry. Q10 Pharmaceutical Quality System

GMP Pharma BV. Netherlands

Making Improvement Work in Pharmaceutical Manufacturing Some Case Studies. Ronald D. Snee

ICH Q7 GMP for Active Pharmaceutical Ingredients

Q8(R2): Pharmaceutical Development

FDA Guidance for Industry Update - Process Validation

PROPOSED UPDATED TEXT FOR WHO GOOD MANUFACTURING PRACTICES FOR PHARMACEUTICAL PRODUCTS: MAIN PRINCIPLES (JANUARY 2013)

Process Validation Guidance Requirements (FDA and EU Annex 15: Qualification and Validation)

Content Uniformity (CU) testing for the 21 st Century: CDER Perspective

Workshop A Design Space (DS)

Fusion Pro QbD-aligned DOE Software

Highly Potent APIs The right platform, people and procedures for successful development and manufacturing

Michael Kickuth and Thomas Friedli

A Preliminary Proposal for a Pharmaceutical Engineering Graduate Program

ICH guideline Q8, Q9 and Q10 - questions and answers volume 4

PRODUCT DEVELOPMENT GUIDE

GENERAL GUIDANCE FOR INSPECTORS ON HOLD-TIME STUDIES

Extended abstract: Model-based computer-aided framework for design of process monitoring and analysis systems

Commercial Manufacturing - Qualification & Validation-related GMP Deficiencies and Other Lifecycle Considerations

Multivariate Tools for Modern Pharmaceutical Control FDA Perspective

Statistical estimation using confidence intervals

Guidance for Industry

Post-Approval Change Management: Challenges and Opportunities An FDA Perspective

White paper: FDA Guidance for Industry Update Process Validation

Process Validation: Practical Aspects of the New FDA Guidance

Innovation and Continuous Improvement in Pharmaceutical Manufacturing

ICH Q 10 Pharmaceutical Quality System PQS DIA China Annual Meeting, May 2010

Microbiology and Auditing. Don Singer

ANDA CHECKLIST FOR CTD or ectd FORMAT FOR COMPLETENESS and ACCEPTABILITY of an APPLICATION FOR FILING

NEW CHEMICAL ENTITIES

Building the Business Case for Automated Rapid Testing: Translating the benefits of rapid automated microbial testing into dollars saved

The problem with waiting time

Simulation-based Optimization Approach to Clinical Trial Supply Chain Management

Big Data to Design Quality

Risk-Based Change Management Using QbD Principles

Design of Experiments for Analytical Method Development and Validation

Stochastic Forecasting of New Product Diffusion With Adherence to Historical Data

GRADUATE CERTIFICATE IN GOOD MANUFACTURING PRACTICE (GMP) Plan your career for tomorrow and you will be ahead of your competitors

State of Control Over the Lifecycle and Process Validation (New and Legacy Products)

Introduction to Enteris BioPharma

2014 Annual Report on Inspections of Establishments

A Stability Program for the Distribution of Drug Products

whitepaper Impact of Formula- on Pharmaceutical Manufacturers

The Geography of Markets for Technology: Evidence from Bio- Pharmaceuticals

Overview of Pre-Approval Inspections

Multivariate Chemometric and Statistic Software Role in Process Analytical Technology

Qualified Persons in the Pharmaceutical Industry Code of Practice 2009, updated August 2015

Welcome to the ChemCon Company Presentation: 6 Steps to Success for our Customers!

Implementing New USP Chapters for Analytical Method Validation

Process Performance Qualification. Demonstrating a High Degree of Assurance in Stage 2 of the Process Validation Lifecycle

PHARMACEUTICAL QUALITY SYSTEM Q10

Is a Career in the. Pharmaceutical. Check out our online Student Center to find out more:

A NUSAGE-PharmEng Pharmaceutical and Biotechnology Training Program

Research needs in pharmaceutical excipients: implications of a global supply chain

2.3 QUALITY OVERALL SUMMARY Sakura Tablet

QbD Considerations for Analytical Methods - FDA Perspective

STABILITY TESTING OF NEW DRUG SUBSTANCES AND PRODUCTS

MONTE CARLO SIMULATION FOR INSURANCE AGENCY CONTINGENT COMMISSION

Annex 6. Guidance on variations to a prequalified product dossier. Preface

Quality Management System (QMS) for Active Pharmaceutical Ingredients (API) Manufacturers

Pharmaceutical Quality Management System: Current Concept

An FDA Perspective on Post- Approval Change Management for PAT and RTRT

Pharmaceutical Engineering: The Lisbon Masters Program

What is Process Validation?

Research Article Prospective Validation of Paracetamol Tablet Dosage Form

Guidance for Industry

Global Lab Capabilities Pharma Biotech

Pharmaceutical Auditor Training

Guidance for Industry

Applying Statistics Recommended by Regulatory Documents

Streamlining clinical trial supply management

Revealing the Big Picture Using Business Process Management

Quality Risk Management The Pharmaceutical Experience Ann O Mahony Quality Assurance Specialist Pfizer Biotech Grange Castle

ICH Q10 and Change Management: Enabling Quality Improvement

Guidance for Industry Quality Systems Approach to Pharmaceutical CGMP Regulations

Step-by-Step Analytical Methods Validation and Protocol in the Quality System Compliance Industry

Assay Development and Method Validation Essentials

Salary Survey Domestic, International Salary Survey and Country Cost Analysis

Pharmaceutical Sciences

EUROPEAN COMMISSION HEALTH AND CONSUMERS DIRECTORATE-GENERAL. EudraLex. The Rules Governing Medicinal Products in the European Union

Risk Based Pre-Approval Inspection

Valentina Gualato, Ph.D. Process Development Scientist

Compliance Control Procedure Execution Management System Reduces Compliance Risks by 10X

Transcription:

Proceedings of the 2007 Crystal Ball User Conference MONTE CARLO SIMULATIONS FOR RISK ANALYSIS IN PHARMACEUTICAL PRODUCT DESIGN Bir Gujral Freeman Stanfield Doug Rufino DSM Pharmaceuticals Inc. 5900 Greenville Blvd Greenville NC 27834 ABSTRACT Risk Analysis is one of the essentials of Process Analytical Technology (PAT) being adopted in the Pharmaceutical Industry as per the new guidance of the FDA. Under PAT, the quality is designed into a Pharmaceutical product, rather than established by testing of a finished batch. The traditional spreadsheet analysis of quality outcomes does not give any probability information on quality. In PAT, the critical product quality variables are identified from the historic data. Crystal Ball software is run on critical parameters to build Monte Carlo simulation models. The simulation model provides probability data for almost all possible outcomes by analyzing the statistics of simulations. The probability of quality in the final product can be predicted with confidence. 1 INTRODUCTION Pharmaceutical development and manufacturing has not changed its fundamental paper-based infrastructure for decades. The regulatory requirements were not very encouraging to make such a change. Until recently, the Pharmaceutical Industry and FDA were not falling into line with respect to utilizing innovations into manufacturing processes to bring the Pharmaceutical sector into the twenty-first century. The US Pharmaceutical Industry currently spends approximately $90 billion on manufacturing (Helfrich, 2005). By improving manufacturing efficiency by 5%, the industry could save over $4.5 billion annually. Leading pharmaceutical companies, generic and contract manufacturing organizations have prioritized programs designed to eliminate the routine, non-value-added tasks through automation. The foremost Pharmaceutical companies are now adopting a new approach to automating compliance by utilizing innovative technologies and building quality into the products. Under the new paradigm, the focus is to reduce risk and to provide higher productivity and improved quality. The automations allow us to manage data across the entire Pharmaceutical Industry-within a plant itself (intraplant) and from plant to plant (interplant). The uncertainty regarding a situation indicates risk, which is the possibility of loss, damage, or any other undesirable event. Low risk strategies translate to a high probability of success, profit, or some form of gain. The 2004 risk-based regulatory approaches under Process Analytical Technology (PAT) recognize the level of scientific understanding of formulations and manufacturing process factors affecting the product and quality performance and the capability of process control strategies to prevent and mitigate the risk of producing a poor quality pharmaceutical product. The objective of PAT is to improve understanding and control the manufacturing process, which is consistent with our current drug quality system: quality cannot be tested into products, it should be built in or it should be by design. The increased emphasis of PAT on building quality into products allows us to have more focus on multi-factorial relationship among raw materials, manufacturing processes, and their impact on quality. This is a basis to understanding the relationship among various critical process and formulation factors and developing effective risk mitigation strategies. A desired goal of PAT is to design and develop processes that will ensure a predefined quality at the end of the manufacturing process. To obtain this target all critical sources of variability are identified and explained; the variability is managed by the process and product quality attributes which can be accurately and reliably predicted over the design space established for the materials used, process parameters and manufacturing conditions. The formulation of a drug involves complex coordination of a variety of physical, chemical, and biological factors and processes (Willis, 2004). Thus, any practical efficiency improvements will incorporate a knowledge base that contains a scientific understanding of how these variables interrelate and a means of applying this knowledge to different formulation scenarios. The design-of-experiment (DOE) evaluations are critical to the success of PAT.

By analyzing current methodologies and results in a systematic and statistical manner, pharmaceutical manufacturers will build a database of practical experiences that will enable them to simulate and test new processes. This informatics-based approach will allow Pharmaceutical companies to identify the variables that are most critical to the final desired product and its behavior, to decide where they need to insert controls into the process, and discover the factors that control sample degradation. 2 CONTENT UNIFORMITY Content Uniformity is a measure of the variations in the amount of active ingredient in individual units comprising a batch (Murphy, 2003). It is not always possible to obtain absolute homogeneous mixing of the drug with the excipients. Factors such as different densities, different particle sizes and different shapes contribute to different settling tendencies and flow characteristics, which causes variations in the content uniformity. In the formulation of Product A, we use API, Dextrate, Silicon Dioxide and Magnesium Stearate. Dextrate acts as excipient or filler and is directly compressible tablet diluents, while Silicon dioxide acts as a glident and Magnesium Stearate is a lubricant. The 500 mg strength Product A is made in batches of 260,000 tablets. We made about 125 batches in 2006, some of which failed because of out of specification Content Uniformity. The blending of active pharmaceutical ingredients (APIs) with various excipients is a common step in the solid dosage form manufacturing process. Typically, the API in the compressed blend is analyzed in the finished dosage form for potency and content uniformity. For existing formulations, we ensure that both the active and excipients are uniformly distributed throughout the solid dosage form. But each pharmaceutical product has a distinctive blend of API and excipients and thus each blending operation is also unique.the homogeneity of the blend is critical in defining the uniformity of dosage units within a batch of tablets, especially in the case of direct compression products. Pharmaceutical Scientists recognize that it is unlikely that content uniformity of the dosages form is achieved unless the blend is mixed to a uniform level (Brush, et al., 2006). In a recent proprietary study, 48% of the variations in manufacturing processes were attributed to blending as a major cause. Blend uniformity is addressed in the Current Good Manufacturing Practices (cgmp) regulations and drug approval programs. Section 211.110 of cgmp requires manufacturers to establish control procedures that include adequacy of mixing to assure uniformity and homogeneity. The regulations, however, do not specify the blend testing approach, nor the particulars as to the acceptance criteria, limits, or methods for the testing. The FDA (61 FR 20103) implied the need for blend uniformity testing of all routine manufacturing batches to make certain that a high level of quality was maintained throughout a process, and not just upon completion of the final product The content uniformity of a Pharmaceutical product is proportional to its blend uniformity. If during blending of raw materials, the blend uniformity is not achieved, the final product is liable to fail in its content uniformity. Product A is more problematic in achieving blend uniformity than most other pharmaceutical products because of various factors chiefly the difference in the particle sizes of API and excipient. But why does most of the Product A, production meet the specifications of content uniformity while only some of total production has failed content uniformity? There are various factors, which are not the same on every batch. These factors are the risk factors causing failed content uniformity of Product A, the risk factors that contribute to failed content uniformity are given below: Particle size differential between API and Dextrates: for API, the largest particle size is of 90 microns while the largest particle size of Dextrates is of 1100 microns The percentage of API in Product A formulation is very high as compared to the percentage of API in the formulations of other Pharmaceutical products The correlations between the particle size of API and excipients are not accounted for The interactions among other parameters of raw materials like moisture content, assay of API are not accounted for The formulations of Product A is not adjusted as per the interactions among the parameters of raw materials The process parameters are not adjusted as per the differences in the particle sizes of API and Dextrates In the Pharmaceutical Industry, drug product quality has largely been based on a combination of locking down inprocess controls to assure the same things are done for each commercial batch, locking down product specifications to assure the same tests are passed for the commercial batches as were passed for the development batches, and GMP to assure the facility is under control. Through the process of creating the PAT policy, this system is challenged by the approach used by industries where the product quality is more directly evident to the consumer. It is apparent that controlling manufacturing variability by controlling a manufacturing process, in real time, is a more reliable approach to assuring product quality when compared to testing the finished product and disposing of the batch if it fails the specification. page 2 of 6

In a dynamic approach to process control, the important process attributes are measured during the process to control it and determine its endpoint. Adjustments can be made to the control variables in order to accommodate any variability in the material entering the operation. This approach of using the manufacturing operation as a lens to focus the materials into the most desired range of properties, greatly reduces product variability. 3 EXPERIMENTAL DATA We collected data of five batches of Product A for its content uniformity and the parameters of the raw materials used in those batches. The data for raw materials used to get content uniformity is given in Table 1 below. Tablet Batch # Table 1: Data of Raw Materials Used to Manufacture Product A Content Uniformity (%) Emedex Particle Size (Average)(um) API Moisture Content (%) API Assay (%) API Particle Size (Average) (um) 1 96.00 461 15.8 97.5 8.8 2 95.10 446 15.3 100.7 7.0 3 94.50 446 15.3 100.7 7.0 4 96.50 446 14.9 100.6 11.0 5 98.00 446 14.9 100.6 11.0 We are focusing on content uniformity of Product A and the content uniformity is affected by particle sizes of API and the excipients. To do the risk analysis, we will build a DOE model based upon the variability of Particle Size of API and Particle Size of excipient to get relationship between content uniformity and the input variables. We will run Monte Carlo simulation on the derived equation to do the forecasting of content uniformity. The Monte Carlo simulation model will also help us to find the impact of each variable, i.e. particle size of API and particle size of excipient on content uniformity. The building of mathematical models is described in the next section.. 4 BUILDING OF DOE MODEL We built a Screening model of DOE by taking Particle Sizes of excipients (PS_1) and Particle Sizes of API (PS_2) as Factors and Content Uniformity (CU) as a Response. Upon fitting the model with the experimental data set, we found that the DOE model is not only affected by particle size of API and excipient but there are interactions between the particle Sizes of API and that of the Particle Sizes of excipients that has a large impact on DOE model. The reliability of impact is observed by adjusted R 2 value of the model. These interactions are captured to get the true relationship between the Content Uniformity and Particle Sizes of API and excipients. In our Screening DOE Model, the value of adjusted R 2 is 0.643 as shown in Figure 1, indicating that this is a viable model. In Table 2, the values of coefficients of Particle Sizes of API, Particle Sizes of excipients helps to build an empirical relationship between Content Uniformity and input variables. page 3 of 6

Investigation: Overview graph of Content Uniformity2.3mip (PLS, comp.=2) Scaled & Centered Coefficients for Content Uniformity 0.50 0.00 % -0.50-1.00 PS_2 PS_1 PS_2*PS_1 N=5 R2 Adj.=0.643 DF=2 Q2=0.660 RSD=0.8078 Conf. lev.=0.95 MODDE 7-3/28/2007 1:28:39 PM Figure 1: Coefficients of Inputs for Content Uniformity as Shown in DOE Model Table 2: The Values for the Coefficients of Inputs for Content Uniformity Derived from DOE Model Content Uniformity Coeff. Std. Err. P Conf. int(±) Constant -306.159 -- -- -- PS_2 45.3593 -- -- -- PS_1 0.889397 -- -- -- PS_2*PS_1-0.100329 -- -- -- The derived Equation from DOE model is: 5 RISK ANALYSIS N = 5 Q2 = 0.660 DF = 2 R2 = -- Comp. = 2 R2 Adj. = 0.643 CU= 0. 889397*PS_1+ 45. 3593 * PS_2-0. 100329 *PS_1 * PS_2 306.159 Risk Analysis is a concept that is applied to deal with uncertainty. In this concept, a probability distribution function is assigned to the unknown variables and then the Monte Carlo simulations are run to determine the combined effect of multiple variables. The seed value of the individual variables is calculated by the probability density definition of each variable. A standard sensitivity study shows us the sensitivity of the resulting improvements from the range of outputs from a single variable. page 4 of 6

The risk analysis approach selects values for independent variables as a function of a probability distribution function for each variable. The independent variable that we have in our case study are the particle size of API and the particle size of excipient. The value of content uniformity is viewed as a probability function rather than a single-valued answer. Thus, for a given data the variability in effectiveness can be viewed as a probability distribution. For a given set of data the simulation model gives maximum and minimum values of content uniformity as 101.71 and 87.37 with a most probable value as 95.59 as per the results shown in the table in Figure 2. If the company specifications for content uniformity are between 80 and 120, then there is 100-percent certainty to get content uniformity within specifications as per the given data for input variables. Figure 2: Forecasting for Content Uniformity Figure 3: Sensitivity Analysis of Forecasting of Content Uniformity page 5 of 6

6 CONCLUSION The application of risk analysis in Pharmaceutical product design requires the setting of the problem definition, constraints and objectives in the proper format to apply the tool. The FDA stresses on building quality into products as per the guidance of its PAT initiative. The quality is designed by two mathematical modeling techniques before it is built during manufacturing. The value of content uniformity is optimized for the Particle Size of API and Particle Size of excipients during Monte Carlo simulations after getting an empirical relationship by building a DOE model. The sensitivity report helps to find out which variable has more impact on output. In our model, both the variables have almost equal impact on the value of content uniformity. To build this model, we had only a limited set of data. A large data set for each variable would improve the mathematical model. REFERENCES Brush, P.J. et.al.: Using MEMS to Control Blending at AstraZeneca; Pharmamanufacturing 2006 FDA, Current Good Manufacturing Practice: Amendment of Certain Requirements for Finished Pharmaceuticals; Proposed Rule, (61 FR 20103) Guidance for industry PAT- A Framework for Innovative Pharmaceutical Development, Manufacturing, and Quality Assurance: US Department of Health and human Services, Pharmaceuticals CGMPs, September 2004. Helfrich, J.P. Analytical Method execution Automation: PAT for Pharmaceutical QC laboratory Operations; American Laboratory, March 2005 Murphy, J.R.: Encyclopedia of biopharmaceutical Statistics: 10.1081/E-EBA-120007424, April 2003. Willis, R.C. Pat Pending; American Chemical Society ISSN 1532-4486, December, 2004 BIOGRAPHIES Dr. Barry Gujral is a Process Analytical Technology (PAT) coordinator and is working to implement PAT at DSM Pharmaceuticals Inc Greenville, NC. He has worked in different fields including Analytical Development, Quality Control, Open Chain Management and Operations. Dr Gujral has obtained his MBA from Duke University, Master of Chemical Engineering from Illinois Institute of Technology, Chicago and Doctorate from Meerut University. His special interests includes: Use of Monte Carlo Simulations to do forecasting and risk analysis in financial value-added indicators and in Pharmaceutical Product Design. Dr. Gujral has a US Patent on Cation-exchangers, has published 44 research papers and is co-author of three books in Chemistry. Dr Gujral is also a co-author of paper, Applications of Process Analytical Technology, published in the November 2006 issue of Current Pharmaceutical Analysis. He has participated in IFPAC-07 and presented a poster on Use of Monte Carl Simulations to account for the Variability in Product Design with his colleague Dr Freeman Stanfield. Barry Gujral can be reached at bir.gujral@dsm.com, Tel # 252-707-7220 Dr. Stanfield holds a Ph.D. in Organic Chemistry from the University of Arizona. He has worked in the Pharmaceutical and Biotech industries since 1988, starting as a post-doctoral at Hoffmann-La Roche in Nutley, NJ. His last 3 positions have been at DSM Pharmaceuticals (Greenville, NC), MedImmune, Inc.(Gaithersburg, MD), and Sigma Genosys Biotechnologies. (The Woodlands, TX). Dr. Stanfield has held postions of increasing responsiblity at DSM Pharmaceuticals over the past seven years. He is now the Manager of Analytical Development, with three Chemistry groups reporting to him. He is married with one daughter. In his spare time Dr. Stanfield enjoys tournament chess, writing poetry, playing guitar, and archery. Doug Rufino is the Sr. Director of QC and Analytical Development at DSM Pharmaceuticals Inc, in Greenville, NC. He has 23 years of experience in the Pharmaceutical Industry covering R&D, QC, Pilot Plant Manufacturing, and QA. He has worked for various organizations such as Warner-Lambert, Bristol-Myers Squibb, and Johnson & Johnson before joining DSM in 2001. He has been utilizing the Lean Six Sigma operational platform for 7 years and is a certified Greenbelt. He heads up the PAT and Lean Six Sigma Programs at DSM. His background is in Microbiology and is a graduate of Rutgers University in New Brunswick, NJ. He is married with four children and enjoys woodworking and recreational scuba diving. page 6 of 6