SlideShare a Scribd company logo
1 of 48
Download to read offline
Boot Camp for FDA CMC/GMP Initiatives 
         for Biotech and Specialty Pharma

         Design Space InPharmatics LLC
         Edward A. Narke
         PDA  Annual Conference
         March 14­17, 2010 – Orlando



1 / 48
Company Information
     Design Space InPharmatics (DSI) is a consulting firm 
     providing integrated CMC drug development and CMC 
     Regulatory services to emerging pharmaceutical and 
     biotechnology companies 
     DSI provides support for its customers from the initial 
     phases of development through successful 
     manufacturing and Regulatory approval




2 / 48
Objectives of the Presentation

    Provide a perspective of the Quality‐by‐Design 
    application for the start‐up to midsize sponsor 
    Provide a perspective on how CTD Module 3 might be 
    constructed in parallel with milestone target planning 
    during Phase development  
    Improve alignment with Regulatory expectations, 
    place greater emphasis on sharing critical scientific 
    information essential to regulatory decision making, 
    and  help enhance the efficiency of the Regulatory 
    process


3 / 48
Outline of the Presentation

    Drivers for QbD
    Why it‘s worth the Effort ‐ Providing Business 
    Benefits
    Incentives and Barriers
    Examples of how it can work




4 / 48
Drivers

Drivers for Quality by Design 
     Prompted by ICH 1, 2
         >   “Assurance of product quality is derived from careful attention to a number of 
             factors including selection of quality parts and materials, adequate product and 
             process design, control of the process, and in‐process and end‐product testing.”
         >   “Due to the complexity of today’s medical products, routine end‐product testing 
             alone often is not sufficient to assure product quality for several reasons.”

     More efficient change control assessment; fewer amendments, 
     reduced testing, easier problem solving, less production 
     downtime, fewer failed batches etc.
     Empirical approach, anecdotal (disconnected) data still 
     acceptable today, however QbD is the future – so be prepared!

1.   ICH Q8, Q9 Q10
2.   Guidance for Industry  PAT — A Framework for Innovative Pharmaceutical Development, Manufacturing, and Quality 
     Assurance 


5 / 48
Drivers

QbD
     Introduced into the CMC review process in 2004 as a result of 
     the pharmaceutical current Good Manufacturing Practice (cGMP) 
     for the 21st Century Initiative
     Expectations outlined in PAT Guidance and ICHQ8, Q9 and Q10
     Proposed implementation comprised of 5 key steps
         >   Identification of Product Attributes of significant importance to 
             safety/efficacy
              >   Quality Target Product Profile (TPPP)
              >   Critical Quality Attributes (CQAs)
         >   Design of the Process to deliver these attributes
         >   A Robust Control Strategy to ensure consistent Process Performance
         >   Validation and Regulatory Filing of the Process
              >   Demonstrating the effect of the Control Strategy

6 / 48
Drivers

QbD
     Familiar Foundation for these activities include:
         >   Risk Assessment
         >   Raw material management
         >   Use of Statistical Approaches
         >   PAT etc.

     Several Case Studies have already been published 
     highlighting benefits
         >   Yu, L.X. et al. (2004) Applications of process analytical technology to crystallization process. Adv. Drug Deliv. Rev. 
             56, 349–369
         >   Gnoth, S. et al. (2007) Process analytical technology (PAT): batch‐to‐batch reproducibility of fermentation 
             processes by robust process operational design and control. J. Biotechnol. 132, 180–186 
         >   Nail, S.L. and Searles, J.A. (2008) Elements of quality by design in development and scale‐up of freeze‐dried 
             parenterals. BioPharm Int. 21 (1), 44–53 
         >   Harms, J. et al. (2008) Defining design space for biotech products: case study of Pichia pastoris fermentation. 
             Biotechnol. Prog. 24, 655–662




7 / 48
Why?

Why it‘s worth the effort
     QbD goal: Use resources in the most efficient manner by 
     producing a better quality data package which will provide 
     flexibility throughout the API/product life cycle
         >   Collect the right data, not necessarily more data*
         >   Data that is information rich, building product knowledge 
         >   Avoid checkbox mentality*
         >   Get the most mileage from experiments throughout 
             development. E.g.., use early data/knowledge base to guide 
             DoE – requires initial hard data as input
         >   Easier to respond to questions from the Agency




8 / 48
Why?

Why it is worth the effort?

     QbD provides a chance to develop new ideas and 
     opportunities to explore new options to meet 
     regulatory requirements and operational 
     flexibility
         >   Gain experience for future Agency expectations
         >   Still collecting data, but in a different manner




9 / 48
Incentives and Barriers

Incentives and Barriers
     Caveats:
      >   Extra effort, DoE in development (Increased $ spent 
          initially during development to define design space = 
          potential returns)
           ‐ Gain Savings depends on project, timing – not guaranteed!
           ‐ Payoff may be delayed until post approval
           ‐ Why does it make good business sense to start a QbD effort when 90% 
             of the candidates fail to make it to market?

      >   Internal fear within companies to push the boundaries / 
          fear of failure and resistance to change – especially near 
          filing time. 



10 / 48
Incentives and Barriers

Incentives and Barriers
Critical Role of Start­ups and First­Product Companies
     Business Perspective
      >   Provide a new pipeline for New Products
      >   Return a profit to stakeholders
     Regulatory Perspective
      >   Address recent initiatives and expectations moving forward
           ‐ Quality by design (QbD) continues to receive much attention. 
           ‐ Although several QbD concepts are being practiced, continued 
             dialogue and partnership between the industry and its 
             regulators will be the key to successful QbD realization



11 / 48
Incentives and Barriers

Incentives and Barriers
     The Facts
     “The degree of sponsor experience with FDA regulations and 
     procedures is generally of importance. Large US­based 
     companies have the highest first­cycle approval rate, at 
     approximately twice the rate of small biotechnology 
     companies with no prior FDA approvals. 
     The underlying drivers seem to be lack of personnel with US 
     regulatory experience and suboptimal­internal regulatory 
     processes’”(1)

1. Independent Evaluation of FDA's First Cycle Review Performance ­ Retrospective Analysis Final Report, Booz, Allen, Hamilton. 
      FDA (January 2006).




12 / 48
Incentives and Barriers

Incentives and Barriers
Shortfall of Start­ups and First­
  Product Companies
     Management in these companies 
     encounter the constant pressure of
      >   Raising funds
                                               CMC              Clin/Nonclinical
      >   Building a creative and effective 
          team
                                               CMC Regulatory      Clinical Research

      >   Keeping ahead of the competition
                                                  Quality 
                                                                     Pharm/Tox
                                                 Assurance
     Focus is on ensuring a viable and 
     effective clinical development            CMC Operations       Medical Affairs


     strategy to keep the product moving 
     forward

13 / 48
Incentives and Barriers

Incentives and Barriers
Consequences of Shortfalls
     In many instances 
      >   Few resources are available to 
          devote to the challenge of 
          developing a viable and effective 
          long­term CMC regulatory strategy    CMC   Clin/Nonclinical




     Lack of focus in this area, coupled 
     with frequent lack of CMC regulatory 
     compliance experience can be a 
     formula for disaster



14 / 48
Incentives and Barriers

Barriers

     Challenges (in the new economy)
      >   Little control on knowledge base for late stage in‐
          licensed, acquired compounds
           ‐ Gaps exist between FIH and POC

      >   Late changes in process, methods etc. that limit the 
          utility of previous data
      >   When using Contract Manufacturing 
           ‐ Gaps occur between vendors
           ‐ Lack of sharing the knowledge between sponsor and CMO
           ‐ Limited resources do not allow for scientific understanding

15 / 48
Incentives and Barriers

Incentives

     Motivation for Business Benefit
      >   Better scientific understanding, easier decisions, (less 
          data and samples to manage?)
      >   Need commitment and better understanding of the 
          benefits to build momentum (QbD is extra work during 
          development, streamlining will be a challenge)
      >   When using Contract Manufacturing 
           ‐ Create an environment of knowledge sharing



16 / 48
How it might Work

Example of how it might work

     Milestone planning
     QbD Blueprint
     Development Phases




17 / 48
How it might Work

Example of how it might work

     Milestone planning – Everyone has a Target
     QbD Blueprint
     Development Phases




18 / 48
Milestone Planning

       CMC‘s Role in the Development Process

                                                          Early Dev              POC                                  Post‐
                         Discovery          Pre‐IND                                              Registration
                                                         (Phase 1/2)          (Phase 2/3)                            Approval

          DISCOVERY                                          NEW CHEMICAL ENTITY PRODUCT DEVELOPMENT
                                                 PRECLINICAL RESEARCH                        CLINICAL STUDIES               NDA REVIEW


Lead Identification                                                                                             Synthesis & Purification
  Lead Optimization                                                                  Manufacturing Formulation/Process Optimization
     Identify Candidate                                                                         Phase 1
       Synthesis & Characterization                                                                       Phase 2
                          Screening and
                              Planning                                                                          Phase 3
                             Definition Phase
                        Pre­Formulation                                                              Accelerated Development/Review
                                                                                                                            Treatment IND

                                                        Animal Testing                                                Parallel Track

                                                                                 Short­Term
                                                                                             Long­Term
                                                       Institutional Review 
                                                                     Boards
                                                                                                   Assays for Therapeutic Efficacy

                 2 ­ 5 years                                   1 ­ 3 years                                   8 ­ 15 years
                                                                             IND submitted

       19 / 48
Milestone Planning

You Must Have a Target!
                                    IND submitted                             NDA submitted

                                                 Early Dev            POC                              Post‐
          Discovery          Pre‐IND                                                  Registration
                                                (Phase 1/2)        (Phase 2/3)                        Approval

                                                                                              Continuous 
                 What is 
What?          correlated
                                                                                              Improvement
                                                              Confirmation 
                to What?                                      Phase = 
                                                              Product 
                                     Optimize Phase =         Knowledge
                                     Building                 •Process 
                                                               Optimization
                                     Robustness
                                                              •Design‐space 
                                     •API Alternate Route      defined
                                      Scouting                •Validation via QbD
                                     •To‐Be Marketed,         •Optimization within 
              Design Phase =          Formulation Design       filied ranges
              Building the           •Site Transfer           •Post‐launch support
              Basics                 •QbD/PAT                 •Life Cycle Strategy
              •API Route scouting
                                     •IVIVC                             And How?
              •API Impurity                          What  
               Identification
              •API Solid Form                       causes
               Selection
              •Initial Risk                         What?
               Assessment
              •Tox Formulation
              •FIH Formulation
20 / 48
Milestone Planning

Management of Targets in the CTD
     Management of Knowledge in Module 3
      >   Provide interwoven experimental evidence that supports a 
          more systematic approach to product development.
      >   Emphasize critical knowledge, analysis and assessment
     Start with standardized templates
      >   Provides framework for most CMC dossier projects
      >   Ensures proper implementation of regulatory strategy in dossier filings
           ‐ Enhances quality and reduces risks in CMC dossier projects
      >   Helps achieve on‐time delivery of NDA/MAA CMC investigational and 
          marketing application filings




21 / 48
Milestone Planning

Manage Knowledge in Module 3

Compliance Data
    API and Product general           Justification of Specification
    properties                        Methods
    Control of starting materials     Validation of methods
    and Excipients
                                      Analytical development history
    Control of reagents, solvents, 
    auxiliary materials               Typical batch data
    Control of intermediates          Reference standards
    Elucidation of API structure      Stability
    List of impurities
    Specifications

22 / 48
Milestone Planning

Manage Knowledge in Module 3

Continuous Conformance Data
    Use CTD Module 3: P2, S26 and S45, P55 as the primary 
    review documents
    Present to regulators all they need to know to make an 
    approval decision
    Present a compelling description and critique of the Quality 
    component in the CTD to make it information and knowledge 
    rich




23 / 48
Milestone Planning

Manage Knowledge in Module 3
    P.2.1 Components of the Drug Product
      >   Which physical chemical characteristics of the drug substance affect drug product development, 
          manufacture, or performance?
      >   What evidence supports compatibility between excipients and the excipients and the drug 
          substance? 


    P.2.2 Drug Product 
      >   What attributes should the drug product possess?
      >   How was the drug product designed to have these attributes? 
      >   What (if any) were alternative formulations or mechanisms that have been investigated?
      >   How were the excipients and their grades selected?
      >   How was the final formulation optimized? 
      >   What are the design space for critical parameters and attributes, and how justified?


    P.2.3 Manufacturing Process Development 
      >   Why was the manufacturing process described in P.3 selected for this drug product?
      >   How are the manufacturing steps (unit operations) related to the drug product quality?
      >   How were the critical process parameters identified, monitored, and/or controlled?
      >   What is the scale­up experience with the unit operations in this process? 
      >   What are the design space for critical parameters and attributes, and how can they be 
          justified?
24 / 48
Milestone Planning

Manage Knowledge in Module 3




25 / 48
Blueprint

Blueprint for how it can work
     Milestone planning
     QbD Blueprint – Creating the Data
      > Build Quality Target Product Profile
      > Identify CQAs
      > Characterize Design Space
      > Identify Control Strategy
      > Validation the Process
      > File the Data

     Development Phases
26 / 48
Blueprint

Blueprint for QbD – An Overview

                 QbD Blueprint
            Build Quality Target Product 
                   Profile (QTPP)
              Identify Critical Quality 
                 Attributes (CQAs)

             Characterize Design Space

             Identify Control Strategy

                Validate the Process

                   File the Data!




27 / 48
QTPP

Build Quality Target Product Profile (QTPP)
QbD Blueprint
                          What would need to be achieved to ensure that quality 
                          affecting safety and efficacy is realized?
 QTPP
                          Start at the End and work backwards!
   Key Questions

   Conditions/Features    Build a ‘‘prospective” summary of the quality characteristics
                                Dosage form and route of administration strength(s) 
   Basis/Considerations
                                Therapeutic moiety release or delivery
   Milestone/Result ‐
   Further Actions
                          Pharmacokinetic characteristics (e.g. dissolution and 
 CQAs                     bioavailability) appropriate to the dosage form
                          Quality criteria (e.g. sterility and purity)
 Design Space

 Control Strategy         QTPP should be established as soon as product has been 
                          identified as a viable candidate for commercialization and 
 Validation               should be revisited at key stages of development
                          Forms the basis of design for the product
 File the Data




28 / 48
CQAs

Identify Critical Quality Attributes (CQAs)
QbD Blueprint

 QTPP                     Desired attributes of a successful Product include?
                          What are the relevant CQAs?
 CQAs

  Key Questions           CQA identification can be performed using risk‐based analysis, in 
                          accordance with the ICH Q9 guidance  
  Conditions/Features     Risk assessment should account for design of the molecule (hot 
                          spots); engineering studies, non‐clinical studies, clinical studies with 
  Basis/ Considerations   the product and/or other platform products; and published 
                          literature
  Milestone/Result ‐
  Further Actions         Attributes known to impact safety and efficacy (such as dissolution, 
                          sterility, solubility or aggregate species) rank high in criticality 
 Design Space             To have no impact on safety and efficacy (such as qualified 
                          impurities) rank low in criticality
 Control Strategy

 Validation               The Critical Attributes used to guide the product and process 
                          development!
 File the Data            Along with QTPP,  forms the basis of Design for the product!




29 / 48
Design Space

Design and Characterize

QbD Blueprint
                    Characterizing the Product Design Space
 QTPP
                         Once CQAs have been identified, the concept of design 
 CQAs                    space can be extended to product quality
                         This product design space should be documented in the 
 Design Space            regulatory filing in the form of in‐process and release 
                         specifications that define the acceptable variability in 
                         CQAs
   Considerations   Characterizing the Process Design Space
                         Process characterization studies can be used to define the 
 Control Strategy
                         acceptable variability in process parameters
                         The approach for defining a process design space has been
 Validation              discussed often in the literature e.g. risk analysis, design
                         of experiments (DOE), linkage of the inputs versus the
 File the Data
                         critical attributes
                         The acceptable ranges for a process should be
                         documented in the regulatory filing.




30 / 48
Control Strategy

Identify Control Strategy
QbD Blueprint            What assures process performance and product quality?
 QTPP                    What are the controls, derived product and process 
                         understanding that assures process and product quality?
 CQAs

 Design Space            IPC testing 
                         Raw material controls
 Control Strategy        Specifications
                         Product characterization
  Key Questions          Container closure system
                         Stability studies
  Basis/Considerations   Validation
                         Comparability studies
  Conditions/Features
                         Will depend of the criticality of the quality attribute and 
  Milestone/Result ‐     capability of the process and analytical methods
  Further Actions
 Validation
                         Using an active control strategy allows manufacturing controls 
 File the Data           to be altered (within the design) to remove or reduce the 
                         variability caused by process inputs
                         QC moved Upstream!


31 / 48
Validation

Validation of Process and Control Strategy
QbD Blueprint

 QTPP
                         Control strategy defined and the product and process design 
 CQAs                    spaces are established
 Design Space            Validation to demonstrate that the process will deliver a product 
                         of acceptable quality if operated within the design space
 Control Strategy
                         Regulatory filing is then compiled, to include Knowledge 
                         Linkages
 Process Validation
                              Acceptable ranges for all key and critical operating 
                              parameters that define the designed process space, in 
   Conditions/Features
                              addition to the more restricted operating space (NOR, 
                              PAR) typically described today
   Milestone/Result ‐
   Further Actions
 File the Data




32 / 48
Reg Filing

 Regulatory Submission
QbD Blueprint            Internal quality system of the manufacturer needs to provide 
                         adequate oversight during QbD implementation to any changes 
 QTPP                    occurring that do not have to pass through regulatory review 
 CQAs
                         and approval
                         Does your Knowledge have linkages?
 Design Space            What does the Agency say?

 Control Strategy

 Process Validation 
 and Filing              Can process improvement during the product life cycle in 
 Regulatory Filing       terms of process consistency and throughput be approved 
                         with reduced submissions?
  Key Questions

  Basis/Considerations
                         With a design space developed and approved under the QbD 
  Conditions/Features    paradigm, process changes within this design space will not 
                         require further review or approval?
  Milestone/Result ‐     Any changes to the design space itself need to be characterized, 
  Further Actions        validated and approved by the regulatory authorities prior to 
                         implementation


 33 / 48
Reg Filing

Examples of a Linkages
     Cross reference between CTD section P2 (Pharm. Development) 
     and relevant sections in S (Drug Substance) and in P (Drug 
     Product)
          ‐ S 1.3 Properties of the active substance
          ‐ S.2 Manufacture
          ‐ S 3.1 Studies on Polymorphism (experimental data)
          ‐ S 4.1 Specifications relating to control of physical forms
          ‐ S.4.3 Analytical methods used
          ‐ S 4.5 Justification of Specifications


          ‐ P 2: Influence of the polymorphic forms on product characteristics – dissolution, 
            stability
          ‐ P 5.1 Product Specifications
          ‐ P 5.6 Justification of Specifications


34 / 48
Blueprint

QbD – Collecting the Data
Key question:   How and when to invest in process science 
                and process characterization to really get 
                to know the manufacturing process and its 
                outcome
Basics:         This investment should be made wisely
                understanding the science does not come cheap 
Marginal conditions:    ‐ Seek out regulatory authority advice 
                        ‐ It’s not about a company believing that it 
                                    knows more about its process and 
                                    product than the regulatory authority 
                        ‐ It’s about involving the regulatory authority 
                                    as a team player
Milestones:             ‐ A reality check for your CMC regulatory   
                                 compliance strategy at any transition 


35 / 48
Development Phases ‐ Overview

A Target for each Milestone
     Milestone planning
     QbD Blueprint
     Development Phase
      >   Pre‐IND
      >   Initiating Phase 1
      >   Early Clin Dev
      >   Clinical Transition from Phase 2 to Phase 3
      >   POC to Registration
      >   Continuous Improvement

36 / 48
Development Phases ‐ Overview


Development Phases ‐ Overview


                   Development Phases

                         Pre‐IND

                     Initiating Phase 1

                       Early Clin Dev

                Transition from Ph 2 to Ph 3

                    POC to Registration

                 Continuous Improvement




37 / 48
Milestone Planning

Critical Role of Integrated Planning
     Not in the Guidances
     Applies to Small Molecules, Biotech and Vaccines
     Most Traditional approach:
      >   Design Phase ‐ Initiating Phase 1 clinical trials 
      >   Optimize Phase ‐ Clinical transition from Phase 2 to Phase 3
      >   Confirm Phase ‐ Submission of the marketing application 

Everyone has a Target!



38 / 48
Development Phases ‐ Overview

The Development Process

                                             Early Dev              POC                                Post‐
     Discovery             Pre‐IND                                                 Registration
                                            (Phase 1/2)          (Phase 2/3)                          Approval
                                              Trials, Scale-up
                                              non-GMP/GMP
          •CMC
          •API Characterization
          •API Route and Scale‐
           up
          •Toxicology/FIH 
           Formulation
          •Definition of QTPP
                                                  Optimize                                             Continuous 
          •ID CQAs                                   Phase           •Process Optimization            Improvement
                                                                     •Design Space 
          •Methods                        •QbD 
           Development Planning and Implementation
                                                                      Validation                   • Life‐Cycle 
                                                                     •TechTransfer/
          •Stability Program
                          Definition Phase•Risk Assessment           •Additional Supply 
                                                                                                     management.
                                           •To‐Be marketed            Chain etc.
                                            Formulation Design
                                                                          Confirmation 
                Design Phase              •Process Design
                                          •Control Strategy                  Phase
          •Clin                           •IVIVC
          •TPP                            •ID CQAs
                                          •Transfer to 
          •Preclin                         Operations
          •Proof of Concept
           •Tox 
           •DDI



39 / 48
Pre‐IND

Pre‐IND Phase 
Development Phases
                          •Are you ready to File the IND?
 Pre­IND                  •Have you defined the Quality Target Product Profile?

   Key Questions

   Conditions/Features    •API Characterization

   Basis/Considerations   •API Route and Scale up

   Milestone/Result ‐     •Toxicology/FIH Formulation Design
   Further Actions
                          •Methods Development
 Initiating Phase 1
                          •Initial Stability Program 
 Early Clin Dev

 Transition Ph2 to Ph3      Items that affect safety and that can trigger a clinical hold can be 
                            determined/ruled out via the QTPP and identifiaction of the 
 POC to Registration        critical attributes!
 Continuous Improve




40 / 48
Phase 1

Initiating Phase 1
Development Phases        How Much? How Controlled? How Tight?

 Pre‐IND
                          Modifications to the method of preparation of the new drug substance and 
 Initiating Phase 1       dosage form, and even changes in the dosage form itself, are likely as the 
                          investigation progresses.
  Key Questions           Emphasis in an initial Phase 1 CMC submission should, therefore, be placed on 
                          providing information that will allow evaluation of the safety of subjects in the 
                          proposed study
  Conditions/Features     The identification of a safety concern or insufficient data to make an evaluation 
                          of safety is the only basis for a clinical hold
  Basis/ Considerations
  Milestone/Result ‐       Products do not lend themselves readily to questions of "how much?", "how 
                           controlled?" and "how tight?" at Phase 1
  Further Actions
                           An effective quality risk management assessment does not simply list all of the 
                           CMC challenges for a product at Phase 1, but prioritizes what the sponsor 
 Early Clin Dev            absolutely needs to do or identify at this early stage of drug development to 
                           address these challenges ...(CQAs?)
 Transition Ph2 to Ph3
                           At Phase I, it is too early to think "test method validation" (instead, think 
 POC to Registration       "suitable for use and scientifically sound") or "manufacturing consistency" 
                           (instead, think "manufacturing control") or "product maximum shelf life" 
 Cont‘ Improvement         (instead, think "stable for the duration of the planned study")




41 / 48
Phase 1/2

Early Clinical Development
                         The manufacturing process is critical to ensure the correct 
Development Phases       composition, Quality, and Safety of the product

 Pre‐IND
                         It becomes more critical to carefully control and record  process 
 Initiating Phase 1      development in conjunction with the appropriate testing to 
                         reproduce a comparable Phase 2 investigational drug
 Early Clin Dev
                         Appropriate equipment and controls should be in place in 
                         manufacturing to ensure that unit operations with safety‐related 
   Considerations        functions (e.g., viral clearance, virus/toxin attenuation. 
 Transition Ph2 to Ph3
                         sterilization) perform their function with a high degree of 
                         assurance. Specific IPC testing may also serve to complement 
                         these functions and can eliminate testing down the chain
 POC to Registration

 Cont‘ Improvement       When evaluating the manufacturing process for early Phase 1&2 
                         investigational drugs, it is of particular importance to begin to 
                         evaluate the process and scale you will need to be at the 
                         confirmation stage
                               Best time to define Design Space is before you are 
                               locked into a ‘process’ for pivotal studies.




42 / 48
Phase 2 to 3

Transition from Phase 2 to Phase 3
Development Phases         Are manufacturing decisions scientifically based on 
                           enhanced knowledge of product performance over a wider 
 Pre‐IND                   range of material attributes, processing options and 
                           process?
 Iniatiating Phase 1
                           Quality by Chance (QbC), is a very risky option
 Early Clin Dev            This investment in QbD should be made wisely
                                  Understanding the science does not come cheap
 Transition Ph 2 to Ph 3          Be selective in how limited time, resources and money are invested
                                  Goal is to increase the level of manufacturing process knowledge and 
  Key Questions                   understanding, not just to generate volumes of scientific data

  Basis/Considerations
                           Want to avoid CMC regulatory compliance delays? Seek out Agency advice. 
                           Regulatory authorities do not expect a company to agree with them in every 
  Conditions/Features      instance, but they do expect thoughtful consideration and a scientifically justified 
                           response.
  Milestone/Result ‐
  Further Actions
                           An effective regulatory compliance strategy is critical at this stage to ensure 
 POC to Registration              All chemistry, manufacturing and controls (CMC) activities necessary for 
                                  eventual market approval are being planned
 Continuous Improve               Any identified CMC impediment should be discussed with the 
                                  regulatory agency



43 / 48
Phase 3 to Registration

POC to Registration
Development Phases

 Pre‐IND
                          Provide enough information to permit the regulatory authority 
 Initiating Phase 1       to determine 
                                Whether the process used in manufacturing the drug and 
 Early Clin Dev                 the controls used to maintain its quality are appropriate 
                                and adequate
 Transition Ph2 to Ph3

 POC to Registration
                          Utilize the knowledge
   Conditions/Features

   Milestone/Result ‐
   Further Actions
 Continuous Improvement




44 / 48
Lifecycle Management

 Continuous Improvement
Development Phases       As the expanded body of manufacturing knowledge occurs 
                         with the contract partner, management should ensure that 
 Pre‐IND
                         continual process improvement is evaluated and given an 
 Initiating Phase 1      appropriate response
                         Facilitate continual improvement: the ICH Q10 mandate for 
 Early Clinical Dev      management
                         Avoid slippage of current Good Manufacturing Practice 
 Transition Ph2 to Ph3   (cGMP) compliance
 POC to Registration
                         Are you capable to building adequate commercial 
 Cont‘ Improvement       inventories?
                         How does market expansion opportunities affect Supply 
  Key Questions          Chain?
  Basis/Considerations
                         The people involved, their competency, training, and 
  Conditions/Features    cooperation, are the key element to the success or failure of 
                         managing a process and product 
  Milestone/Result ‐
  Further Actions



 45 / 48
Summary
     Develop strategies for a QbD approach
      >   These quality initiatives should not be a “one size fits all“ approach to 
          development

     Put together alignment,  guide transition and build 
     a value chain
      >   Strive for seamless Transition and Knowledge Transfer from early 
          Development to launch and beyond
      >   There will always be questions but they should be managed based on the 
          level of risk to quality 

     Enablers – ICH guidance’s Q8, Q9 and Q10 (and 
     soon Q11)
      >   Implementation will take time and experience as well as Open 
          Communication
46 / 48
Acknowledgements

     Anthony Durning
     David Urquhart
     Daniel Torok
     Suzan Aschmies
     Hedley Rees




47 / 48
Thank you …




48 / 48

More Related Content

What's hot

Quality by-Design (QbD) by Mr. Nitin Kadam.
Quality by-Design (QbD) by Mr. Nitin Kadam.Quality by-Design (QbD) by Mr. Nitin Kadam.
Quality by-Design (QbD) by Mr. Nitin Kadam.Nitin Kadam
 
World First 3D Printing | ZipDose® Technology | SPRITAM® | Aprecia | Journal ...
World First 3D Printing | ZipDose® Technology | SPRITAM® | Aprecia | Journal ...World First 3D Printing | ZipDose® Technology | SPRITAM® | Aprecia | Journal ...
World First 3D Printing | ZipDose® Technology | SPRITAM® | Aprecia | Journal ...Navaneethakrishnan Palaniappan
 
QBD Quality by design for Immediate release dosage form
QBD Quality by design for Immediate release dosage formQBD Quality by design for Immediate release dosage form
QBD Quality by design for Immediate release dosage formKushal Saha
 
Quality by design(QBD)
Quality by design(QBD)Quality by design(QBD)
Quality by design(QBD)Balu Khandare
 
QUALITY BY DESIGN PPT BY GIRIJA
QUALITY BY DESIGN PPT BY GIRIJAQUALITY BY DESIGN PPT BY GIRIJA
QUALITY BY DESIGN PPT BY GIRIJAGirija Dandu
 
Quality by design (qbd) a review
Quality by design (qbd) a reviewQuality by design (qbd) a review
Quality by design (qbd) a reviewajaykumarpa
 
Epr 4 09 Qb D Pat
Epr 4 09 Qb D PatEpr 4 09 Qb D Pat
Epr 4 09 Qb D PatPedroNY
 
Implementing a QbD program for linkedin
Implementing a QbD program for linkedinImplementing a QbD program for linkedin
Implementing a QbD program for linkedinSteven (Steve) Laurenz
 
Quality by design
Quality by designQuality by design
Quality by designsuhasini
 
Quality-By-Design In Pharmaceutical Development
Quality-By-Design In Pharmaceutical DevelopmentQuality-By-Design In Pharmaceutical Development
Quality-By-Design In Pharmaceutical DevelopmentShubhangiParbhane1
 
FMEA, DoE & PAT : Three Inseparable Organs of Quality Risk Management (QRM) B...
FMEA, DoE & PAT : Three Inseparable Organs of Quality Risk Management (QRM) B...FMEA, DoE & PAT : Three Inseparable Organs of Quality Risk Management (QRM) B...
FMEA, DoE & PAT : Three Inseparable Organs of Quality Risk Management (QRM) B...Shivang Chaudhary
 
Pharmaceutical Quality by Design (QbD)
Pharmaceutical Quality by Design (QbD)Pharmaceutical Quality by Design (QbD)
Pharmaceutical Quality by Design (QbD)Md. Zakaria Faruki
 

What's hot (20)

Quality by-Design (QbD) by Mr. Nitin Kadam.
Quality by-Design (QbD) by Mr. Nitin Kadam.Quality by-Design (QbD) by Mr. Nitin Kadam.
Quality by-Design (QbD) by Mr. Nitin Kadam.
 
Quality by design (qbd)
Quality by design (qbd)Quality by design (qbd)
Quality by design (qbd)
 
World First 3D Printing | ZipDose® Technology | SPRITAM® | Aprecia | Journal ...
World First 3D Printing | ZipDose® Technology | SPRITAM® | Aprecia | Journal ...World First 3D Printing | ZipDose® Technology | SPRITAM® | Aprecia | Journal ...
World First 3D Printing | ZipDose® Technology | SPRITAM® | Aprecia | Journal ...
 
QBD Quality by design for Immediate release dosage form
QBD Quality by design for Immediate release dosage formQBD Quality by design for Immediate release dosage form
QBD Quality by design for Immediate release dosage form
 
Qbd1
Qbd1Qbd1
Qbd1
 
Quality by design(QBD)
Quality by design(QBD)Quality by design(QBD)
Quality by design(QBD)
 
QUALITY BY DESIGN PPT BY GIRIJA
QUALITY BY DESIGN PPT BY GIRIJAQUALITY BY DESIGN PPT BY GIRIJA
QUALITY BY DESIGN PPT BY GIRIJA
 
Quality by design (qbd) a review
Quality by design (qbd) a reviewQuality by design (qbd) a review
Quality by design (qbd) a review
 
Quality by Design
Quality by DesignQuality by Design
Quality by Design
 
Quality by design
Quality by designQuality by design
Quality by design
 
Epr 4 09 Qb D Pat
Epr 4 09 Qb D PatEpr 4 09 Qb D Pat
Epr 4 09 Qb D Pat
 
Implementing a QbD program for linkedin
Implementing a QbD program for linkedinImplementing a QbD program for linkedin
Implementing a QbD program for linkedin
 
Quality by design
Quality by designQuality by design
Quality by design
 
Quality-By-Design In Pharmaceutical Development
Quality-By-Design In Pharmaceutical DevelopmentQuality-By-Design In Pharmaceutical Development
Quality-By-Design In Pharmaceutical Development
 
FMEA, DoE & PAT : Three Inseparable Organs of Quality Risk Management (QRM) B...
FMEA, DoE & PAT : Three Inseparable Organs of Quality Risk Management (QRM) B...FMEA, DoE & PAT : Three Inseparable Organs of Quality Risk Management (QRM) B...
FMEA, DoE & PAT : Three Inseparable Organs of Quality Risk Management (QRM) B...
 
Pharmaceutical Quality by Design (QbD)
Pharmaceutical Quality by Design (QbD)Pharmaceutical Quality by Design (QbD)
Pharmaceutical Quality by Design (QbD)
 
Quality by Design
Quality by DesignQuality by Design
Quality by Design
 
Quality-by-Design by chattar
Quality-by-Design by chattarQuality-by-Design by chattar
Quality-by-Design by chattar
 
Quality by Design : Quality Target Product Profile & Critical Quality Attrib...
Quality by Design : Quality Target Product  Profile & Critical Quality Attrib...Quality by Design : Quality Target Product  Profile & Critical Quality Attrib...
Quality by Design : Quality Target Product Profile & Critical Quality Attrib...
 
Quality by design
Quality by design Quality by design
Quality by design
 

Similar to Narke Pda Orlando Mar2010 Boot Camp Final

Process Validation & Regulatory Strategies for Fast-track and Breakthrough Th...
Process Validation & Regulatory Strategies for Fast-track and Breakthrough Th...Process Validation & Regulatory Strategies for Fast-track and Breakthrough Th...
Process Validation & Regulatory Strategies for Fast-track and Breakthrough Th...Peter Dellva
 
Pharmaceutical validations- research and reviews journal of pharmaceutical qu...
Pharmaceutical validations- research and reviews journal of pharmaceutical qu...Pharmaceutical validations- research and reviews journal of pharmaceutical qu...
Pharmaceutical validations- research and reviews journal of pharmaceutical qu...M. Luisetto Pharm.D.Spec. Pharmacology
 
Qa tools and techniques
Qa tools and techniquesQa tools and techniques
Qa tools and techniquesprashik shimpi
 
PharmaceuticalQuality by Design (QbD) An Introduction Process Development a...
PharmaceuticalQuality by Design (QbD)   An Introduction Process Development a...PharmaceuticalQuality by Design (QbD)   An Introduction Process Development a...
PharmaceuticalQuality by Design (QbD) An Introduction Process Development a...Bachu Sreekanth
 
Quality by design (qb d).
Quality by design (qb d).Quality by design (qb d).
Quality by design (qb d).sawantanil
 
Pbi consultancy services aug 2012
Pbi consultancy services aug 2012Pbi consultancy services aug 2012
Pbi consultancy services aug 2012MichaelMcNamara
 
Best Practices and Points to Consider for Small-Medium Bio-Pharma Companies
Best Practices and Points to Consider for Small-Medium Bio-Pharma CompaniesBest Practices and Points to Consider for Small-Medium Bio-Pharma Companies
Best Practices and Points to Consider for Small-Medium Bio-Pharma CompaniesCovance
 
Pbi Consultancy Services Aug 2012
Pbi Consultancy Services Aug 2012Pbi Consultancy Services Aug 2012
Pbi Consultancy Services Aug 2012MichaelMcNamara
 
MedAccred Welcome Pack V13 160920
MedAccred Welcome Pack V13 160920MedAccred Welcome Pack V13 160920
MedAccred Welcome Pack V13 160920Adrien Boespflug
 
Six sigma and pharmaceutical industry4
Six sigma and pharmaceutical industry4Six sigma and pharmaceutical industry4
Six sigma and pharmaceutical industry4DrAbdulaziz Saddique
 
Quality assurance tools & techniques
Quality assurance tools & techniquesQuality assurance tools & techniques
Quality assurance tools & techniquesPRANJAY PATIL
 
Does Innovation Pay DIA 2006
Does Innovation Pay DIA 2006Does Innovation Pay DIA 2006
Does Innovation Pay DIA 2006Neil Patel
 
It's easy to turn money into technology...
It's easy to turn money into technology...It's easy to turn money into technology...
It's easy to turn money into technology...Team Consulting Ltd
 
Revised Process Validation
Revised  Process ValidationRevised  Process Validation
Revised Process Validationpharmaakd
 

Similar to Narke Pda Orlando Mar2010 Boot Camp Final (20)

Process Validation & Regulatory Strategies for Fast-track and Breakthrough Th...
Process Validation & Regulatory Strategies for Fast-track and Breakthrough Th...Process Validation & Regulatory Strategies for Fast-track and Breakthrough Th...
Process Validation & Regulatory Strategies for Fast-track and Breakthrough Th...
 
Quality by design
Quality by design Quality by design
Quality by design
 
Pharmaceutical validations- research and reviews journal of pharmaceutical qu...
Pharmaceutical validations- research and reviews journal of pharmaceutical qu...Pharmaceutical validations- research and reviews journal of pharmaceutical qu...
Pharmaceutical validations- research and reviews journal of pharmaceutical qu...
 
Qa tools and techniques
Qa tools and techniquesQa tools and techniques
Qa tools and techniques
 
PharmaceuticalQuality by Design (QbD) An Introduction Process Development a...
PharmaceuticalQuality by Design (QbD)   An Introduction Process Development a...PharmaceuticalQuality by Design (QbD)   An Introduction Process Development a...
PharmaceuticalQuality by Design (QbD) An Introduction Process Development a...
 
Quality by design (qb d).
Quality by design (qb d).Quality by design (qb d).
Quality by design (qb d).
 
Process validation
Process validationProcess validation
Process validation
 
Pbi consultancy services aug 2012
Pbi consultancy services aug 2012Pbi consultancy services aug 2012
Pbi consultancy services aug 2012
 
Best Practices and Points to Consider for Small-Medium Bio-Pharma Companies
Best Practices and Points to Consider for Small-Medium Bio-Pharma CompaniesBest Practices and Points to Consider for Small-Medium Bio-Pharma Companies
Best Practices and Points to Consider for Small-Medium Bio-Pharma Companies
 
concept of validation
concept of validationconcept of validation
concept of validation
 
Pbi Consultancy Services Aug 2012
Pbi Consultancy Services Aug 2012Pbi Consultancy Services Aug 2012
Pbi Consultancy Services Aug 2012
 
MedAccred Welcome Pack V13 160920
MedAccred Welcome Pack V13 160920MedAccred Welcome Pack V13 160920
MedAccred Welcome Pack V13 160920
 
Six sigma and pharmaceutical industry4
Six sigma and pharmaceutical industry4Six sigma and pharmaceutical industry4
Six sigma and pharmaceutical industry4
 
Quality assurance tools & techniques
Quality assurance tools & techniquesQuality assurance tools & techniques
Quality assurance tools & techniques
 
Does Innovation Pay DIA 2006
Does Innovation Pay DIA 2006Does Innovation Pay DIA 2006
Does Innovation Pay DIA 2006
 
It's easy to turn money into technology...
It's easy to turn money into technology...It's easy to turn money into technology...
It's easy to turn money into technology...
 
Revised Process Validation
Revised  Process ValidationRevised  Process Validation
Revised Process Validation
 
Qbd by Anthony Melvin Crasto for API
Qbd by Anthony Melvin Crasto for APIQbd by Anthony Melvin Crasto for API
Qbd by Anthony Melvin Crasto for API
 
801 pdf
801 pdf801 pdf
801 pdf
 
Near Term Planning
Near Term PlanningNear Term Planning
Near Term Planning
 

Narke Pda Orlando Mar2010 Boot Camp Final

  • 1. Boot Camp for FDA CMC/GMP Initiatives  for Biotech and Specialty Pharma Design Space InPharmatics LLC Edward A. Narke PDA  Annual Conference March 14­17, 2010 – Orlando 1 / 48
  • 2. Company Information Design Space InPharmatics (DSI) is a consulting firm  providing integrated CMC drug development and CMC  Regulatory services to emerging pharmaceutical and  biotechnology companies  DSI provides support for its customers from the initial  phases of development through successful  manufacturing and Regulatory approval 2 / 48
  • 3. Objectives of the Presentation Provide a perspective of the Quality‐by‐Design  application for the start‐up to midsize sponsor  Provide a perspective on how CTD Module 3 might be  constructed in parallel with milestone target planning  during Phase development   Improve alignment with Regulatory expectations,  place greater emphasis on sharing critical scientific  information essential to regulatory decision making,  and  help enhance the efficiency of the Regulatory  process 3 / 48
  • 4. Outline of the Presentation Drivers for QbD Why it‘s worth the Effort ‐ Providing Business  Benefits Incentives and Barriers Examples of how it can work 4 / 48
  • 5. Drivers Drivers for Quality by Design  Prompted by ICH 1, 2 > “Assurance of product quality is derived from careful attention to a number of  factors including selection of quality parts and materials, adequate product and  process design, control of the process, and in‐process and end‐product testing.” > “Due to the complexity of today’s medical products, routine end‐product testing  alone often is not sufficient to assure product quality for several reasons.” More efficient change control assessment; fewer amendments,  reduced testing, easier problem solving, less production  downtime, fewer failed batches etc. Empirical approach, anecdotal (disconnected) data still  acceptable today, however QbD is the future – so be prepared! 1. ICH Q8, Q9 Q10 2. Guidance for Industry  PAT — A Framework for Innovative Pharmaceutical Development, Manufacturing, and Quality  Assurance  5 / 48
  • 6. Drivers QbD Introduced into the CMC review process in 2004 as a result of  the pharmaceutical current Good Manufacturing Practice (cGMP)  for the 21st Century Initiative Expectations outlined in PAT Guidance and ICHQ8, Q9 and Q10 Proposed implementation comprised of 5 key steps > Identification of Product Attributes of significant importance to  safety/efficacy > Quality Target Product Profile (TPPP) > Critical Quality Attributes (CQAs) > Design of the Process to deliver these attributes > A Robust Control Strategy to ensure consistent Process Performance > Validation and Regulatory Filing of the Process > Demonstrating the effect of the Control Strategy 6 / 48
  • 7. Drivers QbD Familiar Foundation for these activities include: > Risk Assessment > Raw material management > Use of Statistical Approaches > PAT etc. Several Case Studies have already been published  highlighting benefits > Yu, L.X. et al. (2004) Applications of process analytical technology to crystallization process. Adv. Drug Deliv. Rev.  56, 349–369 > Gnoth, S. et al. (2007) Process analytical technology (PAT): batch‐to‐batch reproducibility of fermentation  processes by robust process operational design and control. J. Biotechnol. 132, 180–186  > Nail, S.L. and Searles, J.A. (2008) Elements of quality by design in development and scale‐up of freeze‐dried  parenterals. BioPharm Int. 21 (1), 44–53  > Harms, J. et al. (2008) Defining design space for biotech products: case study of Pichia pastoris fermentation.  Biotechnol. Prog. 24, 655–662 7 / 48
  • 8. Why? Why it‘s worth the effort QbD goal: Use resources in the most efficient manner by  producing a better quality data package which will provide  flexibility throughout the API/product life cycle > Collect the right data, not necessarily more data* > Data that is information rich, building product knowledge  > Avoid checkbox mentality* > Get the most mileage from experiments throughout  development. E.g.., use early data/knowledge base to guide  DoE – requires initial hard data as input > Easier to respond to questions from the Agency 8 / 48
  • 9. Why? Why it is worth the effort? QbD provides a chance to develop new ideas and  opportunities to explore new options to meet  regulatory requirements and operational  flexibility > Gain experience for future Agency expectations > Still collecting data, but in a different manner 9 / 48
  • 10. Incentives and Barriers Incentives and Barriers Caveats: > Extra effort, DoE in development (Increased $ spent  initially during development to define design space =  potential returns) ‐ Gain Savings depends on project, timing – not guaranteed! ‐ Payoff may be delayed until post approval ‐ Why does it make good business sense to start a QbD effort when 90%  of the candidates fail to make it to market? > Internal fear within companies to push the boundaries /  fear of failure and resistance to change – especially near  filing time.  10 / 48
  • 11. Incentives and Barriers Incentives and Barriers Critical Role of Start­ups and First­Product Companies Business Perspective > Provide a new pipeline for New Products > Return a profit to stakeholders Regulatory Perspective > Address recent initiatives and expectations moving forward ‐ Quality by design (QbD) continues to receive much attention.  ‐ Although several QbD concepts are being practiced, continued  dialogue and partnership between the industry and its  regulators will be the key to successful QbD realization 11 / 48
  • 12. Incentives and Barriers Incentives and Barriers The Facts “The degree of sponsor experience with FDA regulations and  procedures is generally of importance. Large US­based  companies have the highest first­cycle approval rate, at  approximately twice the rate of small biotechnology  companies with no prior FDA approvals.  The underlying drivers seem to be lack of personnel with US  regulatory experience and suboptimal­internal regulatory  processes’”(1) 1. Independent Evaluation of FDA's First Cycle Review Performance ­ Retrospective Analysis Final Report, Booz, Allen, Hamilton.  FDA (January 2006). 12 / 48
  • 13. Incentives and Barriers Incentives and Barriers Shortfall of Start­ups and First­ Product Companies Management in these companies  encounter the constant pressure of > Raising funds CMC Clin/Nonclinical > Building a creative and effective  team CMC Regulatory Clinical Research > Keeping ahead of the competition Quality  Pharm/Tox Assurance Focus is on ensuring a viable and  effective clinical development  CMC Operations Medical Affairs strategy to keep the product moving  forward 13 / 48
  • 14. Incentives and Barriers Incentives and Barriers Consequences of Shortfalls In many instances  > Few resources are available to  devote to the challenge of  developing a viable and effective  long­term CMC regulatory strategy CMC Clin/Nonclinical Lack of focus in this area, coupled  with frequent lack of CMC regulatory  compliance experience can be a  formula for disaster 14 / 48
  • 15. Incentives and Barriers Barriers Challenges (in the new economy) > Little control on knowledge base for late stage in‐ licensed, acquired compounds ‐ Gaps exist between FIH and POC > Late changes in process, methods etc. that limit the  utility of previous data > When using Contract Manufacturing  ‐ Gaps occur between vendors ‐ Lack of sharing the knowledge between sponsor and CMO ‐ Limited resources do not allow for scientific understanding 15 / 48
  • 16. Incentives and Barriers Incentives Motivation for Business Benefit > Better scientific understanding, easier decisions, (less  data and samples to manage?) > Need commitment and better understanding of the  benefits to build momentum (QbD is extra work during  development, streamlining will be a challenge) > When using Contract Manufacturing  ‐ Create an environment of knowledge sharing 16 / 48
  • 17. How it might Work Example of how it might work Milestone planning QbD Blueprint Development Phases 17 / 48
  • 18. How it might Work Example of how it might work Milestone planning – Everyone has a Target QbD Blueprint Development Phases 18 / 48
  • 19. Milestone Planning CMC‘s Role in the Development Process Early Dev  POC Post‐ Discovery Pre‐IND Registration (Phase 1/2) (Phase 2/3) Approval DISCOVERY NEW CHEMICAL ENTITY PRODUCT DEVELOPMENT PRECLINICAL RESEARCH CLINICAL STUDIES NDA REVIEW Lead Identification Synthesis & Purification Lead Optimization Manufacturing Formulation/Process Optimization Identify Candidate Phase 1 Synthesis & Characterization Phase 2 Screening and Planning Phase 3 Definition Phase Pre­Formulation Accelerated Development/Review Treatment IND Animal Testing Parallel Track Short­Term Long­Term Institutional Review  Boards Assays for Therapeutic Efficacy 2 ­ 5 years 1 ­ 3 years 8 ­ 15 years IND submitted 19 / 48
  • 20. Milestone Planning You Must Have a Target! IND submitted NDA submitted Early Dev  POC Post‐ Discovery Pre‐IND Registration (Phase 1/2) (Phase 2/3) Approval Continuous  What is  What? correlated Improvement Confirmation  to What? Phase =  Product  Optimize Phase =  Knowledge Building  •Process  Optimization Robustness •Design‐space  •API Alternate Route  defined Scouting •Validation via QbD •To‐Be Marketed,  •Optimization within  Design Phase =  Formulation Design filied ranges Building the  •Site Transfer •Post‐launch support Basics •QbD/PAT •Life Cycle Strategy •API Route scouting •IVIVC And How? •API Impurity  What   Identification •API Solid Form  causes Selection •Initial Risk  What? Assessment •Tox Formulation •FIH Formulation 20 / 48
  • 21. Milestone Planning Management of Targets in the CTD Management of Knowledge in Module 3 > Provide interwoven experimental evidence that supports a  more systematic approach to product development. > Emphasize critical knowledge, analysis and assessment Start with standardized templates > Provides framework for most CMC dossier projects > Ensures proper implementation of regulatory strategy in dossier filings ‐ Enhances quality and reduces risks in CMC dossier projects > Helps achieve on‐time delivery of NDA/MAA CMC investigational and  marketing application filings 21 / 48
  • 22. Milestone Planning Manage Knowledge in Module 3 Compliance Data API and Product general  Justification of Specification properties Methods Control of starting materials  Validation of methods and Excipients Analytical development history Control of reagents, solvents,  auxiliary materials Typical batch data Control of intermediates Reference standards Elucidation of API structure Stability List of impurities Specifications 22 / 48
  • 23. Milestone Planning Manage Knowledge in Module 3 Continuous Conformance Data Use CTD Module 3: P2, S26 and S45, P55 as the primary  review documents Present to regulators all they need to know to make an  approval decision Present a compelling description and critique of the Quality  component in the CTD to make it information and knowledge  rich 23 / 48
  • 24. Milestone Planning Manage Knowledge in Module 3 P.2.1 Components of the Drug Product > Which physical chemical characteristics of the drug substance affect drug product development,  manufacture, or performance? > What evidence supports compatibility between excipients and the excipients and the drug  substance?  P.2.2 Drug Product  > What attributes should the drug product possess? > How was the drug product designed to have these attributes?  > What (if any) were alternative formulations or mechanisms that have been investigated? > How were the excipients and their grades selected? > How was the final formulation optimized?  > What are the design space for critical parameters and attributes, and how justified? P.2.3 Manufacturing Process Development  > Why was the manufacturing process described in P.3 selected for this drug product? > How are the manufacturing steps (unit operations) related to the drug product quality? > How were the critical process parameters identified, monitored, and/or controlled? > What is the scale­up experience with the unit operations in this process?  > What are the design space for critical parameters and attributes, and how can they be  justified? 24 / 48
  • 26. Blueprint Blueprint for how it can work Milestone planning QbD Blueprint – Creating the Data > Build Quality Target Product Profile > Identify CQAs > Characterize Design Space > Identify Control Strategy > Validation the Process > File the Data Development Phases 26 / 48
  • 27. Blueprint Blueprint for QbD – An Overview QbD Blueprint Build Quality Target Product  Profile (QTPP) Identify Critical Quality  Attributes (CQAs) Characterize Design Space Identify Control Strategy Validate the Process File the Data! 27 / 48
  • 28. QTPP Build Quality Target Product Profile (QTPP) QbD Blueprint What would need to be achieved to ensure that quality  affecting safety and efficacy is realized? QTPP Start at the End and work backwards! Key Questions Conditions/Features Build a ‘‘prospective” summary of the quality characteristics Dosage form and route of administration strength(s)  Basis/Considerations Therapeutic moiety release or delivery Milestone/Result ‐ Further Actions Pharmacokinetic characteristics (e.g. dissolution and  CQAs bioavailability) appropriate to the dosage form Quality criteria (e.g. sterility and purity) Design Space Control Strategy QTPP should be established as soon as product has been  identified as a viable candidate for commercialization and  Validation should be revisited at key stages of development Forms the basis of design for the product File the Data 28 / 48
  • 29. CQAs Identify Critical Quality Attributes (CQAs) QbD Blueprint QTPP Desired attributes of a successful Product include? What are the relevant CQAs? CQAs Key Questions CQA identification can be performed using risk‐based analysis, in  accordance with the ICH Q9 guidance   Conditions/Features Risk assessment should account for design of the molecule (hot  spots); engineering studies, non‐clinical studies, clinical studies with  Basis/ Considerations the product and/or other platform products; and published  literature Milestone/Result ‐ Further Actions Attributes known to impact safety and efficacy (such as dissolution,  sterility, solubility or aggregate species) rank high in criticality  Design Space To have no impact on safety and efficacy (such as qualified  impurities) rank low in criticality Control Strategy Validation The Critical Attributes used to guide the product and process  development! File the Data Along with QTPP,  forms the basis of Design for the product! 29 / 48
  • 30. Design Space Design and Characterize QbD Blueprint Characterizing the Product Design Space QTPP Once CQAs have been identified, the concept of design  CQAs space can be extended to product quality This product design space should be documented in the  Design Space regulatory filing in the form of in‐process and release  specifications that define the acceptable variability in  CQAs Considerations Characterizing the Process Design Space Process characterization studies can be used to define the  Control Strategy acceptable variability in process parameters The approach for defining a process design space has been Validation discussed often in the literature e.g. risk analysis, design of experiments (DOE), linkage of the inputs versus the File the Data critical attributes The acceptable ranges for a process should be documented in the regulatory filing. 30 / 48
  • 31. Control Strategy Identify Control Strategy QbD Blueprint What assures process performance and product quality? QTPP What are the controls, derived product and process  understanding that assures process and product quality? CQAs Design Space IPC testing  Raw material controls Control Strategy Specifications Product characterization Key Questions Container closure system Stability studies Basis/Considerations Validation Comparability studies Conditions/Features Will depend of the criticality of the quality attribute and  Milestone/Result ‐ capability of the process and analytical methods Further Actions Validation Using an active control strategy allows manufacturing controls  File the Data to be altered (within the design) to remove or reduce the  variability caused by process inputs QC moved Upstream! 31 / 48
  • 32. Validation Validation of Process and Control Strategy QbD Blueprint QTPP Control strategy defined and the product and process design  CQAs spaces are established Design Space Validation to demonstrate that the process will deliver a product  of acceptable quality if operated within the design space Control Strategy Regulatory filing is then compiled, to include Knowledge  Linkages Process Validation Acceptable ranges for all key and critical operating  parameters that define the designed process space, in  Conditions/Features addition to the more restricted operating space (NOR,  PAR) typically described today Milestone/Result ‐ Further Actions File the Data 32 / 48
  • 33. Reg Filing Regulatory Submission QbD Blueprint Internal quality system of the manufacturer needs to provide  adequate oversight during QbD implementation to any changes  QTPP occurring that do not have to pass through regulatory review  CQAs and approval Does your Knowledge have linkages? Design Space What does the Agency say? Control Strategy Process Validation  and Filing Can process improvement during the product life cycle in  Regulatory Filing terms of process consistency and throughput be approved  with reduced submissions? Key Questions Basis/Considerations With a design space developed and approved under the QbD  Conditions/Features paradigm, process changes within this design space will not  require further review or approval? Milestone/Result ‐ Any changes to the design space itself need to be characterized,  Further Actions validated and approved by the regulatory authorities prior to  implementation 33 / 48
  • 34. Reg Filing Examples of a Linkages Cross reference between CTD section P2 (Pharm. Development)  and relevant sections in S (Drug Substance) and in P (Drug  Product) ‐ S 1.3 Properties of the active substance ‐ S.2 Manufacture ‐ S 3.1 Studies on Polymorphism (experimental data) ‐ S 4.1 Specifications relating to control of physical forms ‐ S.4.3 Analytical methods used ‐ S 4.5 Justification of Specifications ‐ P 2: Influence of the polymorphic forms on product characteristics – dissolution,  stability ‐ P 5.1 Product Specifications ‐ P 5.6 Justification of Specifications 34 / 48
  • 35. Blueprint QbD – Collecting the Data Key question: How and when to invest in process science  and process characterization to really get  to know the manufacturing process and its  outcome Basics: This investment should be made wisely understanding the science does not come cheap  Marginal conditions: ‐ Seek out regulatory authority advice  ‐ It’s not about a company believing that it  knows more about its process and  product than the regulatory authority  ‐ It’s about involving the regulatory authority  as a team player Milestones: ‐ A reality check for your CMC regulatory    compliance strategy at any transition  35 / 48
  • 36. Development Phases ‐ Overview A Target for each Milestone Milestone planning QbD Blueprint Development Phase > Pre‐IND > Initiating Phase 1 > Early Clin Dev > Clinical Transition from Phase 2 to Phase 3 > POC to Registration > Continuous Improvement 36 / 48
  • 37. Development Phases ‐ Overview Development Phases ‐ Overview Development Phases Pre‐IND Initiating Phase 1 Early Clin Dev Transition from Ph 2 to Ph 3 POC to Registration Continuous Improvement 37 / 48
  • 38. Milestone Planning Critical Role of Integrated Planning Not in the Guidances Applies to Small Molecules, Biotech and Vaccines Most Traditional approach: > Design Phase ‐ Initiating Phase 1 clinical trials  > Optimize Phase ‐ Clinical transition from Phase 2 to Phase 3 > Confirm Phase ‐ Submission of the marketing application  Everyone has a Target! 38 / 48
  • 39. Development Phases ‐ Overview The Development Process Early Dev  POC Post‐ Discovery Pre‐IND Registration (Phase 1/2) (Phase 2/3) Approval Trials, Scale-up non-GMP/GMP •CMC •API Characterization •API Route and Scale‐ up •Toxicology/FIH  Formulation •Definition of QTPP Optimize  Continuous  •ID CQAs Phase •Process Optimization Improvement •Design Space  •Methods  •QbD  Development Planning and Implementation Validation • Life‐Cycle  •TechTransfer/ •Stability Program Definition Phase•Risk Assessment •Additional Supply  management. •To‐Be marketed  Chain etc. Formulation Design Confirmation  Design Phase •Process Design •Control Strategy Phase •Clin •IVIVC •TPP •ID CQAs •Transfer to  •Preclin Operations •Proof of Concept •Tox  •DDI 39 / 48
  • 40. Pre‐IND Pre‐IND Phase  Development Phases •Are you ready to File the IND? Pre­IND •Have you defined the Quality Target Product Profile? Key Questions Conditions/Features •API Characterization Basis/Considerations •API Route and Scale up Milestone/Result ‐ •Toxicology/FIH Formulation Design Further Actions •Methods Development Initiating Phase 1 •Initial Stability Program  Early Clin Dev Transition Ph2 to Ph3 Items that affect safety and that can trigger a clinical hold can be  determined/ruled out via the QTPP and identifiaction of the  POC to Registration critical attributes! Continuous Improve 40 / 48
  • 41. Phase 1 Initiating Phase 1 Development Phases How Much? How Controlled? How Tight? Pre‐IND Modifications to the method of preparation of the new drug substance and  Initiating Phase 1 dosage form, and even changes in the dosage form itself, are likely as the  investigation progresses. Key Questions Emphasis in an initial Phase 1 CMC submission should, therefore, be placed on  providing information that will allow evaluation of the safety of subjects in the  proposed study Conditions/Features The identification of a safety concern or insufficient data to make an evaluation  of safety is the only basis for a clinical hold Basis/ Considerations Milestone/Result ‐ Products do not lend themselves readily to questions of "how much?", "how  controlled?" and "how tight?" at Phase 1 Further Actions An effective quality risk management assessment does not simply list all of the  CMC challenges for a product at Phase 1, but prioritizes what the sponsor  Early Clin Dev absolutely needs to do or identify at this early stage of drug development to  address these challenges ...(CQAs?) Transition Ph2 to Ph3 At Phase I, it is too early to think "test method validation" (instead, think  POC to Registration "suitable for use and scientifically sound") or "manufacturing consistency"  (instead, think "manufacturing control") or "product maximum shelf life"  Cont‘ Improvement (instead, think "stable for the duration of the planned study") 41 / 48
  • 42. Phase 1/2 Early Clinical Development The manufacturing process is critical to ensure the correct  Development Phases composition, Quality, and Safety of the product Pre‐IND It becomes more critical to carefully control and record  process  Initiating Phase 1 development in conjunction with the appropriate testing to  reproduce a comparable Phase 2 investigational drug Early Clin Dev Appropriate equipment and controls should be in place in  manufacturing to ensure that unit operations with safety‐related  Considerations functions (e.g., viral clearance, virus/toxin attenuation.  Transition Ph2 to Ph3 sterilization) perform their function with a high degree of  assurance. Specific IPC testing may also serve to complement  these functions and can eliminate testing down the chain POC to Registration Cont‘ Improvement When evaluating the manufacturing process for early Phase 1&2  investigational drugs, it is of particular importance to begin to  evaluate the process and scale you will need to be at the  confirmation stage Best time to define Design Space is before you are  locked into a ‘process’ for pivotal studies. 42 / 48
  • 43. Phase 2 to 3 Transition from Phase 2 to Phase 3 Development Phases Are manufacturing decisions scientifically based on  enhanced knowledge of product performance over a wider  Pre‐IND range of material attributes, processing options and  process? Iniatiating Phase 1 Quality by Chance (QbC), is a very risky option Early Clin Dev This investment in QbD should be made wisely Understanding the science does not come cheap Transition Ph 2 to Ph 3 Be selective in how limited time, resources and money are invested Goal is to increase the level of manufacturing process knowledge and  Key Questions understanding, not just to generate volumes of scientific data Basis/Considerations Want to avoid CMC regulatory compliance delays? Seek out Agency advice.  Regulatory authorities do not expect a company to agree with them in every  Conditions/Features instance, but they do expect thoughtful consideration and a scientifically justified  response. Milestone/Result ‐ Further Actions An effective regulatory compliance strategy is critical at this stage to ensure  POC to Registration All chemistry, manufacturing and controls (CMC) activities necessary for  eventual market approval are being planned Continuous Improve Any identified CMC impediment should be discussed with the  regulatory agency 43 / 48
  • 44. Phase 3 to Registration POC to Registration Development Phases Pre‐IND Provide enough information to permit the regulatory authority  Initiating Phase 1 to determine  Whether the process used in manufacturing the drug and  Early Clin Dev the controls used to maintain its quality are appropriate  and adequate Transition Ph2 to Ph3 POC to Registration Utilize the knowledge Conditions/Features Milestone/Result ‐ Further Actions Continuous Improvement 44 / 48
  • 45. Lifecycle Management Continuous Improvement Development Phases As the expanded body of manufacturing knowledge occurs  with the contract partner, management should ensure that  Pre‐IND continual process improvement is evaluated and given an  Initiating Phase 1 appropriate response Facilitate continual improvement: the ICH Q10 mandate for  Early Clinical Dev management Avoid slippage of current Good Manufacturing Practice  Transition Ph2 to Ph3 (cGMP) compliance POC to Registration Are you capable to building adequate commercial  Cont‘ Improvement inventories? How does market expansion opportunities affect Supply  Key Questions Chain? Basis/Considerations The people involved, their competency, training, and  Conditions/Features cooperation, are the key element to the success or failure of  managing a process and product  Milestone/Result ‐ Further Actions 45 / 48
  • 46. Summary Develop strategies for a QbD approach > These quality initiatives should not be a “one size fits all“ approach to  development Put together alignment,  guide transition and build  a value chain > Strive for seamless Transition and Knowledge Transfer from early  Development to launch and beyond > There will always be questions but they should be managed based on the  level of risk to quality  Enablers – ICH guidance’s Q8, Q9 and Q10 (and  soon Q11) > Implementation will take time and experience as well as Open  Communication 46 / 48
  • 47. Acknowledgements Anthony Durning David Urquhart Daniel Torok Suzan Aschmies Hedley Rees 47 / 48