1

 

                DEPARTMENT OF HEALTH AND HUMAN SERVICES

 

                      FOOD AND DRUG ADMINISTRATION

 

                CENTER FOR DRUG EVALUATION AND RESEARCH

 

 

 

             ADVISORY COMMITTEE FOR PHARMACEUTICAL SCIENCE

 

 

                              Volume I

 

 

 

 

                          Tuesday, May 3, 2005

 

                               8:30 a.m.

 

 

                CDER Advisory Committee Conference Room

                           5630 Fishers Lane

                          Rockville, Maryland

                                                                 2

 

                        P A R T I C I P A N T S

 

      Charles Cooney, Ph.D., Chair

 

      Hilda F. Scharen, M.S., Executive Secretary

 

      Committee Members:

 

      Patrick P. DeLuca, Ph.D.

      Paul H. Fackler, Ph.D., Industry Representative

      Michael S. Korczynski, Ph.D.

      Gerald P. Migliaccio, Industry Representative

      Kenneth R. Morris, Ph.D.

      Marc Swadener, Ed.D., Consumer Representative

      Cynthia R.D. Selassie, Ph.D.

      Nozer Singpurwalla, Ph.D.

      Jurgen Venitz, M.D., Ph.D.

 

      Special Government Employees:

 

      Carol Gloff, Ph.D.

      Arthur H. Kibbe, Ph.D.

      Thomas P. Layloff, Jr., Ph.D.

      Marvin C. Meyer, Ph.D.

 

      FDA Participants:

 

      Gary Buchler, R.Ph.

      Lucinda Buhse, Ph.D.

      Ajaz Hussain, Ph.D.

      Mehul Mehta, Ph.D.

      Vibhakar Shah, Ph.D.

      Helen Winkle

      Lawrence Yu, Ph.D.

                                                                 3

 

                            C O N T E N T S

                                                              PAGE

 

      Call to Order

                 Charles Cooney, Ph.D., Chair                    5

 

      Conflict of Interest Statement

                Hilda Scharen, M.S., Executive Secretary         5

 

      Introduction to Meeting--OPS Update

                Helen Winkle                                     7

 

      Opening Remarks

                Charles Cooney, Ph.D.                           16

 

      Establishing Drug Release or Dissolution

      Specifications:

 

         Topic Introduction

                Ajaz Hussain, Ph.D.                             18

 

         Dissolution Measurement System: Current State

           and Opportunities for Improvement

                Lucinda Buhse, Ph.D.                            45

 

         Questions by Committee Members                         76

 

         Overview of Guidance Documents

           and Decision Process:

 

         Biopharmaceutics Section

                Mehul Mehta, Ph.D.                              95

 

         Questions by Committee Members                        128

 

         Establishing Dissolution Specifications:

           Current Practice

                Vibhakar Shah, Ph.D.                           138

 

         Questions by Committee Members                        156

 

      Open Public Hearing:

                Will Brown, USP                                162

 

      Questions by Committee Members                           171

                                                                 4

 

                      C O N T E N T S (Continued)

                                                              PAGE

 

      Establishing Drug Release or Dissolution

      Specifications: (Continued)

 

         Factors Impacting Drug Dissolution and

           Absorption:  Current State of Science

                Lawrence Yu, Ph.D.                             179

 

         Questions by Committee Members                        198

 

         Summary of Tactical Plan

                Ajaz Hussain, Ph.D.                            208

 

         Committee Discussion and Recommendations              229

 

      Clinical Pharmacology Subcommittee Report

         (via teleconference)

                Jurgen Venitz, M.D., Ph.D.                     284

 

      Questions by Committee Members                           301

                                                                 5

 

                         P R O C E E D I N G S

 

                             Call to Order

 

                DR. COONEY:  I would like to welcome

 

      everyone to this morning's meeting.  We have an

 

      opportunity for an on-time start.  I am Charles

 

      Cooney, the new chair of this committee.  I am

 

      delighted to welcome everyone here, both the

 

      committee members as well as the guests.  We have,

 

      not surprisingly, a full agenda this morning and we

 

      will begin with addressing the conflict of

 

      interest.

 

                     Conflict of Interest Statement

 

                MS. SCHAREN:  Good morning.  The Food and

 

      Drug Administration has prepared general matters

 

      waivers for the following special government

 

      employees, Charles Cooney, Patrick DeLuca, Carol

 

      Gloff, Arthur Kibbe, Michael Korczynski, Thomas

 

      Layloff, Marvin Meyer, Kenneth Morris, Nozer

 

      Singpurwalla and Jurgen Venitz who are

 

      participating in today's meeting of the

 

      Pharmaceutical Science Advisory Committee to, one,

 

      receive an update from the Clinical Pharmacology

                                                                 6

 

      Subcommittee and, two, discuss and provide comments

 

      on the general topic of establishing drug release

 

      or dissolution specifications.

 

                This meeting is being held by the Center

 

      for Drug Evaluation and Research.  Unlike issues

 

      before a committee in which a particular product is

 

      discussed, issues of broad applicability, such as

 

      the topic of today's meeting, involve many

 

      industrial sponsors and academic institutions.  The

 

      committee members have been screened for their

 

      financial interests as they may apply to the

 

      general topic at hand.  Because general topics

 

      impact so many institutions, it is not practical to

 

      recite all potential conflicts of interest as they

 

      apply to each member.  FDA acknowledges that there

 

      may be potential conflicts of interest but, because

 

      of the general nature of the discussions before the

 

      committee, these potential conflicts are mitigated.

 

                With respect to FDA's invited industry

 

      representatives, we would like to disclose that Dr.

 

      Paul Fackler and Dr. Gerald Migliaccio are

 

      participating in this meeting as non-voting

                                                                 7

 

      industry representatives, acting on behalf of

 

      regulated industry.  Dr. Fackler's and Dr.

 

      Migliaccio's role on this committee is to represent

 

      industry interests in general and not any one

 

      particular company.  Dr. Fackler is employed by

 

      Teva Pharmaceuticals and Dr. Migliaccio is employed

 

      by Pfizer.

 

                In the event that the discussions involve

 

      any other products or firms, not already on the

 

      agenda, for which FDA participants have a financial

 

      interest, the participant's involvement and

 

      exclusion will be noted for the record.  With

 

      respect to all other participants, we ask in in the

 

      interest of fairness that they address any current

 

      or previous financial involvement with any firm

 

      whose product they may wish to comment upon.  Thank

 

      you.

 

                DR. COONEY:  Thank you.  Now Helen Winkle

 

      will provide an update.

 

                  Introduction to Meeting--OPS Update

 

                MS. WINKLE:  Good morning, everyone.  I

 

      would like to welcome all the members of the

                                                                 8

 

      advisory committee and to especially welcome Dr.

 

      Charles Cooney as our new chair of the advisory

 

      committee.  We, at FDA, have worked with Dr. Cooney

 

      as a member of the committee and have really felt

 

      that he has provided a lot of input into the

 

      committee's activities, and feel that working with

 

      him in the next two years as chair is going to be a

 

      very important step for all of us.

 

                Before I talk about the agenda for this

 

      session of the advisory committee, I would like to

 

      talk a little bit about our current focus at the

 

      agency or what we are calling a paradigm shift.  I

 

      think it is important for all of us to understand

 

      clearly the changes that we are making in the

 

      agency and the role of the advisory committee in

 

      assisting us in making these changes.  Based on

 

      recent initiatives in FDA, including the

 

      Pharmaceutical cGMP Initiative for the 21st

 

      Century, the PAT Initiative and the Critical Path

 

      Initiative, you can see the shift in FDA's thinking

 

      about regulating product quality.

 

                Specifically, there is a focus in these

                                                                 9

 

      initiatives to place more responsibility on

 

      industry to ensure the quality of their

 

      pharmaceutical products rather than rely solely on

 

      regulatory scrutiny to maintain that quality.  This

 

      is really the paradigm shift, a sharing of

 

      responsibility for drug quality with emphasis

 

      placed on industry to understand their processes

 

      and the underlying science of those processes.

 

                Why would we want to make that change?

 

      There is no evidence that the products out there on

 

      the market are bad products.  There is no evidence

 

      that the agency has done a bad job in serving as a

 

      surrogate for ensuring good quality products for

 

      the consumer.  And, there is no evidence that

 

      industry is not focused on quality as an important

 

      attribute to manufacturing products.  However,

 

      times are changing.  As we enter the 21st century

 

      we have an excellent opportunity to begin to

 

      prepare for how we will handle pharmaceutical

 

      regulation in the future.  The time is ripe for us

 

      at FDA to invest in that future and to ensure that

 

      the direction we are going in is adequate to handle

                                                                10

 

      the changing world of pharmaceutical development

 

      and manufacturing while we continue to be able to

 

      serve the consumer.  It is the right time too to

 

      ensure that our regulatory involvement does not

 

      hinder the innovation and continuous improvement in

 

      manufacturing and ensuring the quality of

 

      pharmaceutical products.

 

                So, FDA has begun a journey towards this

 

      paradigm shift.  I want to say it is a long

 

      journey.  It started several years ago but we have

 

      a long way to go, and we have numerous challenges

 

      along that way.  However, with these challenges

 

      come opportunities and I think this is the

 

      important thing for us and the advisory committee

 

      to remember, that we need to take advantage of

 

      these opportunities.  It is important not only to

 

      take advantage of the opportunities to help us

 

      improve on how we regulate product quality, but

 

      also to ensure that we provide for modernization

 

      both at FDA and within industry for the 21st

 

      century.

 

                The guiding principles of the

                                                                11

 

      Pharmaceutical cGMP for the 21st Century, which

 

      include risk-based orientation, science-based

 

      policies and standards, integrated quality systems

 

      orientation, international cooperation and strong

 

      public health protection, serve to help us in

 

      developing the pathway to restructure the oversight

 

      of the pharmaceutical quality.  As each of you

 

      knows, there are a number of forks in that path and

 

      you, as members of this advisory committee, are

 

      really here to help us determine the right path in

 

      the road to go from a scientific perspective, and

 

      to help and advise us on how to fill the gaps which

 

      exist in the FDA.  These include gaps in

 

      organization, gaps in science and gaps in policy.

 

                The committee has already participated in

 

      discussions on a number of scientific issues which

 

      have helped in formulating a strategy for

 

      addressing many of the questions that have emerged

 

      as a result of this paradigm shift.  We have

 

      already discussed a number of issues which have

 

      significance as we develop our future regulatory

 

      paradigm, including such issues as polymorphism,

                                                                12

 

      bio-inequivalence of generic products, and we have

 

      worked together to support such initiatives as the

 

      Process Analytical Technology Initiative.  The

 

      committee has also been extremely helpful in moving

 

      toward this new paradigm with other discussions

 

      that we have had on various topics.  However,

 

      again, the journey has only just begun.

 

                The agenda for the next two days was

 

      developed to provide an opportunity to discuss two

 

      other scientific topics which are important to us

 

      to better understand and manage in order to move us

 

      steadily along the path of change.  The first topic

 

      is on establishing drug release or dissolution

 

      specifications.  Obviously, how we set

 

      specifications is important to the future.  As we

 

      move to the desired state of pharmaceutical quality

 

      we want to ensure that specifications are based on

 

      mechanistic understanding of how product and

 

      process factors impact product performance.  We are

 

      currently in the process of developing a tactical

 

      plan for setting dissolution specifications.  As

 

      you will hear from the presentations today, we have

                                                                13

 

      developed the fundamentals for this plan which

 

      include a systems view of setting specifications,

 

      ensuring that all factors which affect dissolution

 

      are considered; basically ensuring that we connect

 

      all the dots in CMC to ensure a more comprehensive

 

      and systematic way of setting specifications.

 

                FDA recently held a specifications

 

      workshop in co-sponsorship with the Product Quality

 

      Research Institute.  The workshop indicated a need

 

      for additional efforts to move toward better

 

      setting of specifications in general.  Some of the

 

      specific points that were brought out at the

 

      workshop included a lack of globalization on how

 

      specifications are set; a need to better define

 

      what we should do versus what we can do; a need to

 

      better define the role of the compendia in the new

 

      paradigm; and a need to revisit the decision trees

 

      in ICH Q6A.

 

                Our discussion at the advisory committee

 

      today is not designed to address all the workshop

 

      issues and concerns on setting specifications.  The

 

      discussion today will, however, help us finalize

                                                                14

 

      the tactical plan for setting dissolution

 

      specification and will lay the foundation for our

 

      thinking in setting specifications for CMC and

 

      addressing the specific issues that were identified

 

      at the workshop.

 

                We would appreciate the committee's

 

      comments and suggestions as to what data is needed

 

      to support our plans.  This would include looking

 

      at statistical methodology, etc., and how we might

 

      improve on our thinking in our tactical plan and

 

      specifications setting in general.

 

                At this meeting we will also discuss, as

 

      our second big topic, quality by design and

 

      pharmaceutical equivalence.  As you will remember,

 

      at the last meeting we set the stage in our

 

      discussions on bioinequivalence and bioequivalence

 

      testing of locally acting GI drugs.  At this

 

      meeting our goal is to modernize our general

 

      thinking about pharmaceutical equivalence and to

 

      explore how quality by design can be leveraged to

 

      ensure more rational approaches to decision-making

 

      so that we can move from a reactive environment to

                                                                15

 

      a proactive regulatory scheme of assessing

 

      equivalence.

 

                We will discuss a number of relevant

 

      topics, including biopharm. classification system;

 

      using product development information to address

 

      highly variable drugs; and we will revisit the

 

      concept of decision trees for ensuring a rational

 

      approach to determining bioequivalence for topical

 

      drug products.  We look forward to the committee's

 

      feedback on these extremely important topics, and

 

      that discussion is for tomorrow.

 

                There are a number of other topics we will

 

      cover at this meeting, including an update from the

 

      working group on parametric tolerance interval test

 

      for dose content uniformity.  Bob O'Neill will give

 

      that update.  And an update from the Clinical

 

      Pharmacology Subcommittee.  We will also discuss

 

      with the committee our perceived need to establish

 

      a working group for the review and assessment of

 

      OPS' research programs.  Our goal in looking at our

 

      research programs is to ensure a common approach to

 

      all laboratory work and to ensure that our research

                                                                16

 

      aligns with our overall mission.

 

                Now that we have two laboratories in OPS,

 

      a biotech. laboratory and a lab focused on small

 

      molecules, it is extremely important that this

 

      alignment takes place and we really look forward to

 

      your input on how we can better align these two

 

      laboratories.

 

                As you can see, we have a full agenda but

 

      I think the topics to be discussed are really of

 

      great interest to us as we move down the path to

 

      the desired state for pharmaceutical quality, and I

 

      look forward to a very interesting discussion on

 

      each of these topics.  Thank you.

 

                            Opening Remarks

 

                DR. COONEY:  Thank you, Helen.  I would

 

      like to just add a couple of comments, if I may, to

 

      get us started.  I am delighted to have the

 

      opportunity to work with the FDA and to work with

 

      this committee during the coming two years.  It is

 

      a particularly exciting time because as we look

 

      forward, as Helen has indicated, there are very

 

      important new initiatives on the table with the

                                                                17

 

      cGMP Initiative and the Critical Path Initiative

 

      and these, indeed, lay a foundation that we all

 

      need to work within.  In fact, it is an exciting

 

      opportunity to work within those initiatives to

 

      look at how we can better address some of the

 

      challenges going forward.

 

                Certainly, as we look forward there are

 

      more challenges than there have been in the past.

 

      We are facing a world of increasing molecular

 

      complexity; a world of increasing demands by

 

      consumers; a world in which we have increasing

 

      complexity not just in the molecules but in the

 

      delivery formats of these products and the role of

 

      this committee is very important in helping to

 

      provide advice to the FDA and to the division to

 

      deal with these problems.  I must say, I applaud

 

      the forward-looking and the proactive stance that

 

      is being taken on these issues.

 

                We have some challenging goals today and

 

      tomorrow, not the least of which, of course, is to

 

      stay on time.  But the reason that the challenge of

 

      staying on time is a challenge is because of the

                                                                18

 

      very high content of material that we need to deal

 

      with.  So, I will do my best to try and keep us

 

      within the proper boundaries.

 

                Again, I look forward to working with

 

      everyone.  This will be a very interesting two days

 

      and I see it as an important step in what will be a

 

      continuing series of discussions and activities and

 

      recommendations that we will need to work on with

 

      the FDA.  With that, the first presentation and

 

      discussion this morning will be by Ajaz Hussain,

 

      and we will begin by digging in to establishing

 

      drug release and dissolution specifications.  Ajaz,

 

      please?

 

                Establishing Drug Release or Dissolution

 

                   Specifications Topic Introduction

 

                DR. HUSSAIN:  Thank you, Dr. Cooney.  I

 

      think topic one is entitled quality by design

 

      approach for quality control and assurance of

 

      dissolution rate.  In the background packet, as

 

      well as in the presentations, I have tried to keep

 

      the terminology dissolution rate all along to

 

      illustrate the one challenge which we will not be

                                                                19

 

      discussing today, and that is the metrics for

 

      dissolution rate itself.  We express dissolution

 

      rate as a Q factor which tends to be confounded

 

      with the variability of the assay itself.  So, that

 

      is not the topic for discussion today but I just

 

      wanted to alert you on why the word "rate" keeps

 

      coming back and back again.  So.

 

                Topic one:  Our goal is to seek your

 

      recommendations and endorsement of the proposed

 

      regulatory tactical plan.  With this tactical plan

 

      we hope to start moving towards putting together a

 

      set of regulatory tools and policies that will help

 

      us define elements and details of the elements

 

      necessary to realize the goals of quality by

 

      design.

 

                The question that we are posing to you is

 

      are the tactical steps outlined consistent with the

 

      goals that we have shared with you?  What initial

 

      steps and/or changes would you recommend to improve

 

      this plan?  What additional scientific evidence do

 

      you feel would be necessary to support the

 

      development and implementation of this plan? 

                                                                20

 

      General considerations for identifying and

 

      developing statistical procedures and, in

 

      particular, I want to emphasize for this discussion

 

      today that we have left out a discussion on

 

      statistical procedures because our experience has

 

      been that if we start with that as a topic it leads

 

      to protracted debate, and you will hear one report

 

      on that debate from Bob O'Neill tomorrow, the

 

      debate on parametric tolerance interval that has

 

      been going on for years and hasn't come to any

 

      resolution.  We feel that if you approach it from

 

      scientific foundations first, statistics is simply

 

      a tool to implement the scientific decision

 

      framework.  So, that is the reason we have kept

 

      that out of discussion for today.  Clearly, we are

 

      seeking your specific recommendations and other

 

      recommendations that you may have including how we

 

      should prioritize our work to develop this tactical

 

      plan to a full proposal, which we hope to bring to

 

      you at a subsequent meeting.

 

                What are the proposed steps?  The proposed

 

      steps are to develop an alternate regulatory

                                                                21

 

      approach to demonstrate the suitability of

 

      dissolution measurements system; introduce and

 

      utilize the concept of reproducibility and

 

      repeatability study using the actual pharmaceutical

 

      product for which we set specification.  Here, the

 

      proposal is to consider using the pivotal clinical

 

      lot or the bio. lot as a basis for identifying how

 

      sensitive, or lack of it, it is to a dissolution

 

      test method and estimate the variability in the

 

      method and, therefore, of the product.

 

                So, the first two steps are sort of

 

      together.  But the next three steps are also sort

 

      of in one clump.  We want to move towards a

 

      system-based decision tree for establishing

 

      dissolution rate specification.  Within that

 

      framework I think we would like to utilize

 

      opportunities to utilize the PAT approach for

 

      controlling dissolution rate and development of

 

      real-time quality assurance strategies.  Also, a

 

      decision tree for design-based concepts articulated

 

      in the draft ICH Q8 guideline, which is in your

 

      background packet.

 

                So, those are the decision trees which we

 

      would like to develop.  At the same time, when we

 

      come back with the full proposal to you, we would

                                                                22

 

      like to bring to you a side-by-side comparison of

 

      new and generic drugs because we think this is an

 

      opportunity for both sides, and some of the

 

      frustrations the generic industry feels can be

 

      addressed with this proposal, and I will explain

 

      that towards the end of the day, and explain why

 

      the level of quality assurance or quality control

 

      confidence in the proposed approach will be higher

 

      than what is achieved in the current system.  There

 

      is no doubt in my mind but, clearly, you have to

 

      agree with that.

 

                We also seek today your recommendations on

 

      how we should approach the statistical aspect of

 

      this and then what will help you to discuss this

 

      proposal when it comes to you.  So please give us

 

      your recommendations on how we should prepare a

 

      detailed proposal for a subsequent ACPS meeting.

 

                The other step that I think is important

 

      and is very timely, because this Friday or this

                                                                23

 

      Saturday I leave for Brussels for our ICH meeting,

 

      is that we do intend to seek the discussion here,

 

      and utilize the discussion here, to seek

 

      harmonization of the approach we are proposing

 

      under the ICH, especially the ICH Q8 Part 2, and we

 

      will start developing that guideline in Brussels

 

      next week.

 

                ICH Q8 draft guideline essentially brought

 

      a basis for getting and considering pharmaceutical

 

      development information in a structured way for

 

      pharmaceutical decision-making in the CMC arena at

 

      FDA.  The guideline was constructed with this

 

      figure, on your right, in mind.  You have to focus

 

      your design efforts on the intended use of the

 

      product, the patient population and so forth, that

 

      leads to a product design and that product design

 

      dictates the design specification, which are

 

      customer requirements, and these requirements, some

 

      of them if they are critical, become regulatory

 

      specifications.  Then the product design and design

 

      specification dictates or leads to design of a

 

      manufacturing process to reliably and predictably

                                                                24

 

      deliver those specifications back to deliver the

 

      intended use.

 

                In a systematic way, if you approach

 

      pharmaceutical development in a structured way, you

 

      get some benefit, we believe.  You achieve a higher

 

      degree of process understanding and give regulators

 

      high confidence of low risk of releasing a poor

 

      quality product; high efficiency through continuous

 

      learning and improvement.  And, I think it helps us

 

      to address some of the gaps we have in the current

 

      system.  I have tried to illustrate the current

 

      process within FDA and the manufacturing and R&D

 

      process within industry.  Research and development

 

      will develop the products, transfer them to

 

      manufacturing and then we have, by law, a separate

 

      quality unit to maintain quality assurance.  You

 

      have all the specification results and you have

 

      products which don't have all the specification

 

      results.

 

                In approaching and assessing the quality

 

      we bring a team approach, a multi-disciplinary team

 

      approach which includes pharmacology, toxicology,

                                                                25

 

      CMC review, clin. pharm., bio. pharm. review and

 

      the clinical assessment.  And the decision

 

      collectively is a risk-benefit decision that leads

 

      to an approval of a product.  The approved product

 

      is then transferred--it is called technology

 

      transfer--and the process is validated.

 

                The validation process includes

 

      qualification criteria and so forth, but there is

 

      an element of that which is process qualification.

 

      Process qualification is essentially, in my

 

      opinion, the interaction between materials and

 

      equipment and environment that you really have to

 

      study.  In the current state that essentially is

 

      judged on your ability to repeat it three times.

 

      Since the pharmaceutical development information is

 

      not available for CMC reviewers, the quality of

 

      that and the understanding containing that is not

 

      well understood in the regulatory sense.  So, we

 

      are losing an opportunity to make more rational

 

      decisions.

 

                Now, we have a divide, an organizational

 

      divide within the agency between, say, review and

                                                                26

 

      GMP inspection.  The cGMP process is helping

 

      bridge.  The PAT is an example of how we have

 

      bridged it.  Our experience or learning from the

 

      initiative clearly identified a need for a quality

 

      system orientation.  I would be wrong if I said

 

      that I really did not understand what this really

 

      meant or really didn't care about what quality

 

      system issues were because I was looking from

 

      several years ago.  But I think I have gained a

 

      much deeper understanding of the importance of the

 

      quality system orientation.

 

                So, here is a representation of that from

 

      a paper that we published on innovation and

 

      continuous improvement in pharmaceutical

 

      manufacturing.  Say what you do, do what you say,

 

      prove it and improve it are the elements that make

 

      up a quality system.  Consider the way what you do

 

      is your application to FDA.  So, that is a CMC

 

      review assessment process.  Now, do what you say

 

      can be considered as are you able to manufacture to

 

      the commitments that you have placed in your

 

      application?  Now, there is a gap since our

                                                                27

 

      reviewers don't have an idea that they really do

 

      what they say.  That is a GMP function so there is

 

      uncertainty there.  And prove it.  How do you prove

 

      it?  I think metrics for proving it could be

 

      process capability and the recalls, and this and

 

      that.  If you are unable to prove it you need to

 

      have a collective action and a preventive action.

 

                Our experience has suggested that in most

 

      cases root cause is unknown or a poor analyst is

 

      blamed.  So, we actually don't get to a root cause

 

      generally.  Does the current system support or

 

      facilitate getting to the root cause?  I think that

 

      is the question.  In many ways I think say what you

 

      do and do what you say, if you take that ratio is

 

      process understanding and your ability to prove.

 

      So, in many ways I think you have to think about

 

      that.

 

                Now, a modern quality system has a

 

      dimension of improvement, continuous improvement

 

      and innovation.  The dotted line simply says that

 

      is an option that should be available for industry

 

      to do.  It is dotted because that is not a

                                                                28

 

      requirement per se, but the rest are all

 

      requirements.  So, I think we are trying to address

 

      some of these gaps along the way.

 

                Now, the definition of continuous

 

      improvement is interesting and it really sets the

 

      stage for this discussion.  I have taken the

 

      definition from QS-9000 to illustrate the challenge

 

      we face for continuous improvement.  For those

 

      product characteristics and process parameters that

 

      can be validated using variable data, that is

 

      continuous data, continuous improvement means

 

      optimizing the characteristics and parameters at a

 

      target value and reducing variation around the

 

      target value.  So, in a sense, you need a target

 

      value and you need to have an estimate of variation

 

      to start thinking of continuous improvement.  In

 

      our specification setting often we don't even have

 

      a target value.  So.  And forget variation.  So.

 

                But the second bullet is more important.

 

      For those product characteristics and process

 

      parameters that can be only evaluated using

 

      attribute data, pass/fail, continuous improvement

                                                                29

 

      is not possible until characteristics are

 

      conforming.  If attribute data results do not equal

 

      zero defect it is by definition a non-conforming

 

      product.  Improvements made in these situations are

 

      by definition corrective actions, not continuous

 

      improvement.  And, we have clearly distinguished

 

      between corrective action, which is a risky

 

      scenario, and continuous improvement, which can be

 

      managed differently.

 

                Continuous improvement in processes that

 

      have demonstrated stability, acceptability,

 

      capability and performance--continuous improvement

 

      really is only possible for those products that

 

      have demonstrated stability.  Process validation

 

      today does not give us the assurance that the

 

      process is stable.  So, that is another element.

 

      Acceptable capability, we don't have an estimate of

 

      the capability value.

 

                Now, the reason for finding this out is

 

      that I think we don't use compendial methods as

 

      release specifications.  Actually, the compendium

 

      approach to specifications is right.  That is the

                                                                30

 

      way they should be.  There is nothing wrong with

 

      this specification criteria for the market

 

      standard.  It is perfectly all right.  But it is,

 

      as Janet Woodcock says in her paper, different from

 

      release specification and that is the

 

      distinguishing feature that I think is the problem

 

      here.  If you use market standard as release

 

      specification, then you have all the elements that

 

      hold back continuous improvement.  So, you really

 

      need to distinguish between standards and

 

      specifications.  Unfortunately, in the current

 

      paradigm specifications equals standard.  So, what

 

      we are moving towards is a control strategy that

 

      will allow you to have your market standard but

 

      then have a control philosophy that allows you a

 

      risk-based decision process.

 

                A recent proposal from USP I think is a

 

      step in the right direction.  It is essentially a

 

      similar proposal to the parametric tolerance

 

      interval test to take dissolution specification

 

      criteria towards more of a tolerance interval

 

      approach.  But as you will hear tomorrow from the

                                                                31

 

      parametric tolerance interval discussion, you

 

      cannot approach it as hypothesis testing for every

 

      product batch, and that is one of the discussions

 

      that we will have.  And, there are many challenges

 

      before we even can get to that, and that is a part

 

      of this discussion.  We believe one has to start

 

      with a pharmaceutical science discussion before

 

      developing appropriate statistical tools.

 

                One other challenge for continuous

 

      improvement is the mind set--and this is a major

 

      challenge not only within the U.S. but

 

      globally--that corrective actions is the only way

 

      to force improvement of quality on industry.  This

 

      is direct current paste from the paper that we

 

      issued.  Some would argue that corrective actions

 

      provide the necessary constancy of purpose for

 

      improvement, necessary since manufacturing is a

 

      stepchild of industry because the difference

 

      between cost of manufacturing and price of drugs is

 

      large.  Keeping the system of corrective action

 

      provides the leverage for ensuring improvement, to

 

      ensure the cGMP.

 

                That is a fundamental challenge.  How do

 

      we achieve that?  If you improve your manufacturing

 

      process by reducing variability your regulatory

                                                                32

 

      acceptance criteria will be narrow.  So, that takes

 

      things into a way for continuous improvement.  So,

 

      that is another challenge that we will start

 

      addressing.

 

                The argument has some validity but it is

 

      based on an assumption that current practices,

 

      including measurement systems and product

 

      specification, provide efficient means for

 

      identifying, understanding and then reducing

 

      variability.  For quality assurance in the 21st

 

      century we need a sound basis to verify such

 

      assumptions in the current system.

 

                To emphasize this point further, we

 

      discussed the case of dissolution and that is what

 

      we present to you today.  Let me illustrate an

 

      example, a real case example.  This is an example

 

      of an approved and validated manufacturing process

 

      at a major pharmaceutical company.  I will read the

 

      middle portion of this.  This is the warning

                                                                33

 

      letter:  There is no assurance that the production

 

      in process control procedures established--this is

 

      controlled-release product--to produce a product

 

      that has the quality it is purported to have or

 

      represented to possess.  How did we approve it?

 

      How was it validated?  So, this is after the fact.

 

      The duration of each coating cycle is determined by

 

      the pan operator but is based on visual

 

      determination that the coating solutions are evenly

 

      distributed before proceeding to the next step.  It

 

      is noted that literally 50 percent of the batches

 

      are thrown out every year because of dissolution

 

      failures, and then you have partial release

 

      occurring too.  Doesn't this undermine the entire

 

      credibility of our system?  And, this was

 

      catastrophic for the company.

 

                Now, inability to resolve our

 

      specification observations I think undermines the

 

      credibility of our decision system.  It raises

 

      questions of adequacy of the current decision

 

      system.  It increases the risk of releasing an

 

      unacceptable quality product to the consumer, and

                                                                34

 

      contributes to low efficiency.

 

                Now, corrective action, preventive

 

      action--there are some challenges.  There are

 

      difficult questions faced by manufacturing groups

 

      and regulators since we have a calibrated system

 

      that we use for dissolution and a calibrated system

 

      is a tablet similar to any other tablet that we

 

      use, and the quality is an issue there.  If you

 

      choose to use a calibrated tablet for gauge R&R

 

      study, reproducibility and repeatability study,

 

      what you see there is that the calibrator

 

      variability and its manufacturing process is

 

      confounded within that system.  I am not going to

 

      go through the equations but it is simple algebra.

 

                In addition, we have another challenge.

 

      The challenge is that the assumption of independent

 

      variable cannot be really verified because the

 

      hydrodynamics of the vessels are such--I see our

 

      colleagues from Health Canada here who have been

 

      criticizing this for a long time.  Thank you for

 

      coming, sir.  So, how representative is the

 

      suitability for that product is an issue.

 

                But the need for improvement is not

 

      limited.  We need to be confident of our analysis,

 

      of surveillance samples, consumer complaints, other

                                                                35

 

      investigations.  One of the frustrating jobs that I

 

      have is where we get consumer complaints; we do

 

      investigations; we do dissolution--no answers.  I

 

      mean, you really don't get to the root cause.

 

                I think the basic philosophy that Walter

 

      Shewhart sort of proclaimed years ago is very

 

      important.  Pure and applied science have gradually

 

      pushed further and further the requirements of

 

      accuracy and precision.  However, applied science,

 

      particularly in the mass production of

 

      interchangeable parts, is even more exacting than

 

      pure science in certain matters of accuracy and

 

      precision.  That is the basis of this discussion.

 

                Is the current approach to calibration

 

      adequate?  Dr. Cindy Buhse will share with you her

 

      challenges--as one of the premier labs, probably

 

      the world standard for dissolution at FDA and

 

      elsewhere, and Tom Layloff had started some of

 

      these processes and he is here too--dissolution

                                                                36

 

      testing of the USP wants to require diligent

 

      attention to details, mechanical and chemical.

 

      Dosage forms can respond definitely to small

 

      variations; large differences in dissolution

 

      results are possible unless all parameters are

 

      carefully controlled.  Differences in

 

      reproducibility can often be traced to improper

 

      mechanical calibration or degassing.  Much of that

 

      is mechanical.  When you only have suitability

 

      criteria just based on a tablet, it hides some of

 

      this variability.

 

                We had a rude awakening to this ourselves.

 

      This is really when I started realizing the

 

      confounding nature of the problem that we have.

 

      Just to illustrate how frustrating this experience

 

      was, our marines were contracting malaria when they

 

      were in Liberia and we were asked to see whether

 

      this was a quality problem.  We faced significant

 

      challenges in analysis because I had insisted that

 

      two labs would do this because this was a grave

 

      situation.  Unexpected inter-laboratory differences

 

      highlighted limitations of current calibration. 

                                                                37

 

      Here is just a quote from our DPA lab:  We are at a

 

      loss to explain the difference between DPA and the

 

      Philadelphia district office initial results.  Then

 

      we started tracing it back.  It had to be

 

      mechanical differences and degassing.

 

                Well, I think that is not the only issue.

 

      I think the bigger issue that we are confronted

 

      with is that we need to better understand the

 

      sources of variability in product performance and

 

      quality so as to establish the most appropriate

 

      design specifications for the product that support

 

      continuous improvement and address the increasing

 

      complexity of product designs.

 

                This is another concern.  We are moving

 

      towards drug eluting, towards nano materials,

 

      towards other complex devices and, yet, we don't

 

      have good measurement systems for these products.

 

      We want measurement systems for products intended

 

      for non-oral administration and non-oral drug

 

      delivery systems; develop and implement globally

 

      harmonized proactive regulatory decision system,

 

      including Q6A and Q8.

 

                I just want to sort of lay the foundation

 

      for other aspects that Mehul and Vibhakar will

 

      share with you.  Pharmaceutical development and

                                                                38

 

      dissolution specification without pharmaceutical

 

      development information creates more challenges.

 

      Decisions focus only on dissolution test data.

 

      Tests are often used for both in-process control

 

      and final product testing.  Decision

 

      characteristics focused only on the mean value will

 

      deal with variability indirectly.  Variability

 

      managed indirectly using "disconnecting test

 

      conditions" and acceptance criteria leads to

 

      deterministic interpretation of specifications and

 

      ignores background variability and, as Dr. Woodcock

 

      has said, we need to move towards a probabilistic

 

      decision system.  Specifications are standards and

 

      standards don't give any room for uncertainty or

 

      risk-based decisions.  If you don't meet the

 

      standards, you are off the market.  It is as simple

 

      as that.  So.  And you have event trees as opposed

 

      to decision trees.  It is difficult to resolve

 

      specification observations which could be related

                                                                39

 

      to how we set specifications, and post-approval

 

      changes and optimization in continuous improvement

 

      is difficult.

 

                This is simply an illustration of the gap

 

      that we base all of our decisions on test-to-test

 

      comparison, in vivo to in vitro, and there is an

 

      opportunity to use the design information to make

 

      rational decisions.  Just to illustrate this, again

 

      this is from Health Canada which has been very

 

      proactive and pushing this agenda and I am sorry we

 

      just didn't react more quickly--here is an

 

      illustration of the false-positive and

 

      false-negatives that you get.  The reference

 

      product dissolves 95 percent in 15 minutes, and the

 

      reference AUC, Cmax.  But if you look at product F,

 

      it dissolves very slowly in vitro but, yet, in vivo

 

      it meets the criteria--it is almost identical to

 

      that.

 

                So, there is a formulation attribute that

 

      does this.  For example, if you have a large amount

 

      of organic or insoluble excipient it is a

 

      hydrodynamic effect.  That doesn't happen in vivo. 

                                                                40

 

      The in vivo media, the surface tension, the

 

      hydrodynamics are completely different.  So, you

 

      tend to see this but you also get false-positives

 

      and false-negatives.  If I look at product C, it

 

      has only 62 percent dissolution compared to product

 

      F and has half the Cmax.

 

                There are other differences in how we

 

      approach specification setting.  The difference

 

      between the U.S. and Japan--we included a paper of

 

      the Japanese perspective on this in your background

 

      packet.  Because of the new restrictions I took the

 

      names off.  I had to go back and erase those.  This

 

      is a published paper so I was surprised that I

 

      needed to take the names off.

 

                The point here is this, all are basic

 

      drugs and this is a rule of thumb that has been

 

      known for 30 years, if you have a drug with PK

 

      between 4-6 the best media to illustrate in vivo

 

      performance is that of the PK value.  That is where

 

      the dissolution is slower.  So, the Japanese have

 

      been in that direction.  All our specifications use

 

      0.1 normal, here.  Is that important?  Well, the

                                                                41

 

      Japanese think so because they are very concerned

 

      with hypoacidity in the subjects.  If I really look

 

      at it, with antacids and H2 blockers most of us are

 

      hypoacidic too.  So, is this a gap that we need to

 

      fill is the question that I think we will address

 

      as we go along.  So, you can see the dramatic

 

      difference in dissolution as pH 1.2 to pH 7.2 and

 

      the resulting blood levels.

 

                So, in a sense, the opportunity we are

 

      trying to realize is ICH Q6A actually had it quite

 

      nicely captured in this quote:  The quality of a

 

      drug substance and drug product is determined by

 

      the design development, in-process controls, GMP

 

      controls, process validation and by specifications

 

      applied throughout development and manufacture.

 

      So, you have the goal; you have the decision

 

      characteristics; and you have the life cycle.  The

 

      design development was the missing element in our

 

      decision characteristics.  Now we have an

 

      opportunity to use it more effectively.

 

                This is how ICH Q8 captured that

 

      opportunity, to bring the development and design

                                                                42

 

      information not only to ask the right question but

 

      also to realize the opportunities of flexibility

 

      that might bring.  So, design and development

 

      should impact positively on how we set

 

      specifications in process controls and have more

 

      confidence in process validation and GMP controls.

 

                With that as a background and the reason

 

      for this topic for discussion, in many ways the

 

      tactical plan is an attempt now to go back ten

 

      years and to see how we can do better with our new

 

      information that could come through the PAT process

 

      and the ICH Q8 process.  In many ways we are

 

      reexamining the SUPAC guideline, the dissolution

 

      guideline for '97, the biopharmaceutics

 

      classification assumed in ICH Q6A.  The vector for

 

      the desired state is that we are adding another

 

      layer of variability assessment, identification

 

      assessment and utilization of variability in our

 

      decision-making.  So, the basic fundamental is that

 

      the quality of decisions can only be better so the

 

      current system is the minimum level of quality that

 

      we achieve.

 

                So, for the discussion today Cindy Buhse

 

      will share with you her proposal on measurement

 

      system, how mechanical calibration will be better

                                                                43

 

      and that is what we want to use.  Mehul Mehta will

 

      share with you the general overview of our decision

 

      process in our guidances.  Lawrence Yu is one of

 

      the leading experts I think in sort of modeling

 

      dissolution and in vivo absorption.  So, I have

 

      asked him to share a perspective on the current

 

      state of science.  Then I will come back and

 

      outline the steps of the proposal.  I have a number

 

      of slides in your packet but I will not be using

 

      those slides.  I will be using only the first 16

 

      slides to give you ample opportunity for

 

      discussion.  Those are backup slides.  If there are

 

      questions I will come back to them.

 

                In your background packet I specifically

 

      identified one person by name for his

 

      contributions, and that is Dr. Vinotcha [ph.].  I

 

      think the work he has done in particular--the

 

      reason I am pointing him out today is because he

 

      has decided to retire and I want to recognize his

                                                                44

 

      contribution to dissolution.  He has brought it to

 

      this level and I think taking it beyond that, and I

 

      thank him for that and he is here today.  A number

 

      of people are there from DPA who are experts in

 

      this and I will recognize them at some other point.

 

                With that, I will stop and invite Cindy to

 

      share her thoughts with you.  Any questions before

 

      I leave?

 

                DR. COONEY:  Thank you, Ajaz.  We will

 

      certainly have time for extensive discussion later

 

      but I think, particularly since we are right on

 

      schedule, if anyone has any questions for Ajaz

 

      right now, particularly for clarification of any of

 

      the points he has made, this would be a very

 

      appropriate time to take a moment for this.  Ken?

 

                DR. MORRIS:  Yes, just one quick point on

 

      an early slide where you were talking about the

 

      development process, it actually goes from the

 

      intended use through to development.  I would just

 

      say for clarification, because this is something

 

      that I get quite a lot, what we really want to get

 

      across I think is the idea that when you have the

                                                                45

 

      intended use and the characteristics you really

 

      select your process first.

 

                DR. HUSSAIN:  Yes.

 

                DR. MORRIS:  And then come back to the

 

      formulation.  So, it doesn't necessarily change the

 

      order but it adds a level because that is a

 

      constant source of confusion, particularly when you

 

      are talking about building in dissolution

 

      characteristics.

 

                    Dissolution Measurement System:

 

            Current State and Opportunities for Improvement

 

                DR. BUHSE:  Thank you, Ajaz.  It is going

 

      to be my job to tell everybody a little bit about

 

      dissolution.  Some of you, I know, are very

 

      familiar with it but some of you may never have

 

      experienced it or seen it done and it is kind of a

 

      very different way of testing so I am going to show

 

      you a little bit about the different apparatus you

 

      can choose to do dissolution testing; talk a little

 

      bit about how we currently determine instrument

 

      suitability in terms of calibration, both

 

      mechanical and chemical; and also validation of

                                                                46

 

      dissolution of test methods and what we typically

 

      see in our lab when we take a look at method

 

      validation packages.  Then I am going to show you

 

      some sources of variability within dissolution,

 

      show you examples of how some formulations are

 

      sensitive to some parameters and some formulations

 

      are sensitive to others and we really need to

 

      understand for your particular formulation where

 

      your sources of variability are coming from.  Then

 

      I will just briefly talk about some opportunities

 

      for improvement, many of which Ajaz already alluded

 

      to in his talk.

 

                If you go to USP, there are seven

 

      different dissolution apparatus listed.  They are

 

      all up here.  You can see that the ones I am going

 

      to talk about today mostly are apparatus 1 and 2

 

      because those are the two that are used the most by

 

      most pharmaceutical companies.  We do see some of

 

      the other apparatus occasionally.  Apparatus 3,

 

      reciprocating cylinder, can also be set up for

 

      apparatus 7 so those are actually the same piece of

 

      equipment.  The flow-through cell is used more in

                                                                47

 

      Europe than it is in the United States.  We don't

 

      see much with that here.  Then, apparatus 5 and 6

 

      are used for transdermal delivery systems and they

 

      are actually a modification of apparatus 1 and 2.

 

                What I am going to talk about most today

 

      is apparatus 1 and 2, which is actually the same

 

      piece of equipment and what you are doing is you

 

      are changing the shaft on the different vessels to

 

      change it from apparatus 1 to apparatus 2.

 

      Actually shown in the picture there is apparatus 2

 

      and you can see there are paddles above each one of

 

      the about 900 ml vessels there.  The way

 

      dissolution works is that you are actually testing

 

      6 tablets at once.  I think Ajaz showed that in the

 

      specifications there usually is a specification

 

      which says 6 tablets have to have a certain

 

      dissolution value and if one of those 6 fails you

 

      go to 12 tablets and then you go to 24.  So, you

 

      start with just 6 and if everything goes right,

 

      then you will be done after the 6 tablets.

 

                So, you essentially have 6 different

 

      pieces of apparatus here because each one of those

                                                                48

 

      vessels acts independently.  You would fill each

 

      one with whatever media it is that you want to test

 

      in, whether it is 0.1 normal HCL or water or

 

      simulated intestinal fluid.  There are all sorts of

 

      ranges of media that people use.  So, you put

 

      500-900 ml in these vessels and then for apparatus

 

      2 you just lower the paddle down and start it going

 

      at whatever rpm you decide.  Certainly, that is

 

      another variable you can manipulate.  Then you drop

 

      your tablet or capsule in and then you take a

 

      sample out of the media at whatever time point your

 

      specification is.  If your specification may be 80

 

      percent dissolved after an hour, then after an hour

 

      you would withdraw a small portion of the media and

 

      then you would determine how much the drug has

 

      dissolved.  Usually the determinative step there is

 

      HPLC.  So, you do that for all 6 of these vessels

 

      and then, hopefully, everything dissolves in the

 

      right amount of time and you will be done.

 

                the basket--similar.  You just change the

 

      shaft and you put a basket on and you actually put

 

      the drug in the basket and then you lower the

                                                                49

 

      basket and start it spinning and you go through the

 

      same procedure.

 

                Just so you can see what it looks like,

 

      this shows you what apparatus 3 looks like, which

 

      you can also turn into apparatus 7 by changing the

 

      holders.  You actually would put the tablet or

 

      capsule inside each one of those up at the top.

 

      What it does, it comes up and down inside each one

 

      of these little vessels down at the bottom.  What

 

      you can do with this apparatus is you can change

 

      the media so in every row you can put a different

 

      medium if you want.  So, if you want to start your

 

      capsule dissolving at 0.1 normal HCL and move it to

 

      simulated intestinal fluid, in the first row you

 

      could put acid.  In the next row you could put

 

      intestinal fluid.  In the next row you could put

 

      whatever you want.  Then you can move this

 

      apparatus up, you know make it go up and down for

 

      an hour in one and then move to the next and go up

 

      and down.  So, that is how you could do it with

 

      apparatus 3.

 

                This is apparatus 4, and I think I

                                                                50

 

      mentioned we don't see a lot of this one.  This is

 

      a flow-through cell.  You can see over there, on

 

      the far side, that is what the actual cell looks

 

      like.  So, if you had a capsule or tablet that

 

      didn't completely disintegrate you could put it in

 

      this cell and actually flow through, somewhat like

 

      actually happens in humans--flow through a media

 

      and change it as you go.  You can either recycle it

 

      around or you can actually have a one pass through

 

      media as well and then analyze the media as it is

 

      coming out to see how much drug is dissolved.  For

 

      this one there is also a bunch of different cells,

 

      different geometries that you could put in this.  I

 

      kind of show examples of that there.

 

                Most of what I am going to talk about

 

      today is apparatus 1 and 2, and that is because

 

      that is the majority of what we see in methods that

 

      are given to us for method validation.  When they

 

      use apparatus 1 or 2 they use the USP criteria for

 

      setting up the equipment and for calibrating the

 

      equipment, and I will go over what those parameters

 

      are.  Then, as I think Ajaz said, most tablets and

                                                                51

 

      capsules have a one point acceptance criteria.  For

 

      immediate-release products we see anywhere from 2

 

      to 4 time points, maybe 1 hour, 4 hours, 8 hours,

 

      24 hours depending on the product.

 

                The first thing you are going to do if you

 

      have one of these apparatus, you are going to run a

 

      test method.  You need to ensure that you have

 

      instrument suitability.  The first point I have up

 

      there is which one of these 7 instruments you are

 

      going to use.  What we find is that most people use

 

      1 and 2.  Most people believe that that is what the

 

      FDA wants to see.  I have been to many different

 

      dissolution conferences and, you know, consultants

 

      and companies will get up there and say if at all

 

      possible use apparatus 1 or 2 because that is what

 

      the FDA wants.  I have heard many people say that

 

      so a lot of people try to use 1 and 2.

 

                Then, once you have chosen your

 

      instrument, you need to make sure it is set up

 

      properly for mechanical calibration.  You can see

 

      by the picture that if your shaft is not quite

 

      centered, or if your vessel is not quite seated

                                                                52

 

      right, your rpm aren't calibrated, etc., you can

 

      imagine that you can get different hydrodynamics

 

      from vessel to vessel or from time to time.  You

 

      need to really carefully make sure that everything

 

      is set up properly.  Then, once you have everything

 

      set up properly, you can then run a calibrator

 

      tablet provided by the USP to see if you get within

 

      the range that the calibrator tablet says you

 

      should get.  Then that gives you some measure of

 

      confidence that perhaps you have set this thing up

 

      properly with mechanical calibration.  I think Ajaz

 

      has mentioned that the calibrator tablets actually

 

      are U.S. phenomena and they are not used either in

 

      the European or Japanese pharmacopeias.

 

                Once you have instruments all set up, then

 

      you can certainly do method development/method

 

      validation, and I will talk a little bit about what

 

      we see and what is actually given to us, as the

 

      agency, when it comes to validating the dissolution

 

      method.

 

                Here is an example of some of the

 

      mechanical calibration parameters out of the USP. 

                                                                53

 

      Some of them have specific values.  For instance,

 

      the shaft has to be 2 mm from the centerline, which

 

      means you actually have a 4 mm spread because you

 

      can have one direction and then it spins around to

 

      the other.  You can see there are other parameters

 

      which don't really have any hard numbers associated

 

      with them, such as the wobble--no significant

 

      wobble and that is kind of nebulous there, or no

 

      significant vibration.  So, those are the some of

 

      the USP criteria for setting up the basket and

 

      paddle methods.

 

                The actual calibrator tablets--actually,

 

      our lab in St. Louis had a lot to do with

 

      calibrator tablets coming into being.  It is

 

      certainly the current 10 mg one that is used today.

 

      But they came around in the 1970s and there are two

 

      different calibrator tablets.  One is

 

      disintegrating and one is non-disintegrating.  So,

 

      one pretty much falls apart when it goes into the

 

      dissolution apparatus; the other stays together as

 

      a tablet throughout the calibration procedure.

 

                In 1997 a 50 mg prednisone tablet, the

                                                                54

 

      disintegrating one, was replaced with a 10 mg

 

      tablet which was manufactured at the University of

 

      Maryland, here, and was based on the formulation of

 

      a product that our lab had found was sensitive to a

 

      lot of the parameters of calibration, including

 

      degassing and mechanical calibration, so we thought

 

      it would be a good calibrator tablet.

 

                Actually, last year the working group at

 

      the USP was actually looking for a replacement for

 

      the 10 mg tablet.  It does have quite a bit of

 

      variability associated with it and some stability

 

      issues so they would like to see if they can find

 

      something else.

 

                So, if you are actually calibrating your

 

      apparatus what you would do, if you use your

 

      equipment for both basket and paddle which is what

 

      we do in our lab--a lot of pharmaceutical companies

 

      will have one that will always stay paddle and

 

      another will always stay basket but we go back and

 

      forth.  If you are using the same instrument for

 

      both paddle and basket, what you would do is you

 

      would do 4 different calibration runs.  You would

                                                                55

 

      do both calibrators with the paddle installed and

 

      then you would turn around and do both calibrators

 

      with the basket installed to make sure that your

 

      instrument is set up properly.

 

                How often do you do these?  In our lab we

 

      do it every 6 months.  We do the calibration using

 

      the prednisone 10 mg tablet.  Here is the actual

 

      data on the current lots of calibrator tablets.

 

      The O lot, which has been in effect now for almost

 

      two years I think--you can see there are different

 

      dissolution criteria depending on whether you are

 

      running it in the basket or the paddle method.  You

 

      see there is a fairly wide range.  You can see that

 

      for the basket as long as you are anywhere between

 

      53-77 percent for each vessel you are going to pass

 

      calibration.  So, you have your 6 vessels and this

 

      one, over here, can be 53 and this one, over here,

 

      could be 77 but you are still going to pass

 

      calibration.  Actually, late last year they changed

 

      the ranges of the prednisone tablet because there

 

      were stability issues and a lot of failures in the

 

      market, and you can see that the range is even

                                                                56

 

      wider now, 51-81 percent.

 

                I have also included up there the values

 

      we get in our lab for at least the prednisone

 

      tablet.  For the basket method we get 72.6.  You

 

      can see we run very much on the high end of that

 

      range.  In fact, we do quite often fall out on the

 

      high end.  You can see we tend to run on the low

 

      end of the range on the paddle method for these

 

      calibrator tablets.

 

                The salicylic acid tablet has a much

 

      narrower range.  It is also much less sensitive to

 

      many of the parameters that you set for dissolution

 

      testing so it is not sensitive to degassing; it is

 

      not sensitive to mechanical calibration setup.

 

                The problem often with running these

 

      calibrator tablets is if you do get an out of

 

      specification value, then what do you do?  You

 

      check your mechanical calibration.  It can be

 

      difficult to decide whether the issue is the actual

 

      calibrator tablet itself or the issue is some way

 

      that you set up the instrumentation.

 

                The other problem with the calibrator

                                                                57

 

      tablet is that you can see it has a fairly wide

 

      range.  It can often interfere with a continuous

 

      improvement process.  If your vessels can be

 

      anywhere from 51-81 percent and you are still

 

      passing, what does that say when you are running

 

      your own product and you want to try to narrow down

 

      the variability of your product?  You don't have

 

      much room here I guess to try to keep everything

 

      consistent.

 

                I am going to talk just a little bit about

 

      development and validation.  We don't see a lot of

 

      development data but we do see the validation data

 

      in our lab.  Obviously, when you are developing a

 

      dissolution method you have to decide about all

 

      these different parameters, a lot of which I have

 

      alluded to, and you want to develop a method that

 

      is going to be discriminatory.  You want to be able

 

      to tell between good product and bad product.  You

 

      want the method to be repeatable.  You would like

 

      the method to give you the same results no matter

 

      which lab you are running it in.  I think Ajaz said

 

      we had some trouble with the malaria drug in trying

                                                                58

 

      to get two different labs get the same results.

 

      You have to decide which instrument to use.  Like I

 

      said, most people try to pick 1 and 2 if at all

 

      possible; then what media to run it in.  A lot of

 

      the test methods we get either are in 0.1 normal or

 

      HCL; a lot of them are just plain water.  Then you

 

      have to decide whether degassing is going to be

 

      important or not for your product; and decide

 

      whether or not you need sinkers.  Some products

 

      don't automatically go to the bottom of the vessel

 

      if you are using the paddle method.  You can buy

 

      commercial sinkers, which are these little devices

 

      that you put the tablet in that will actually make

 

      it fall to the bottom, or you can just wrap a wire

 

      around, which is what is in the USP, to make it go

 

      down to the bottom.

 

                Once you have decided all these

 

      parameters, you still need a determinative step,

 

      and that is what the main focus of validation is

 

      for most companies.  So, when we get validation

 

      packages in from companies on their dissolution

 

      test methods, their validation really focuses on

                                                                59

 

      the determinative step.  They do a lot of work on

 

      varying the parameters on the HPLC method but less

 

      data do we see on varying the parameters on the

 

      actual dissolution method.  So, we see more on the

 

      determinative step and less on the actual

 

      parameters that are associated with the dissolution

 

      apparatus.

 

                You can see that there are a lot of places

 

      here where variability can be introduced, and

 

      certainly when developing a product if you want to

 

      have a test method that is going to allow you to

 

      continuously improve your product you really need

 

      to understand what all the sources of variability

 

      are going to be.

 

                This is one of Ajaz's slides.  I think he

 

      showed a similar one earlier which is basically a

 

      slide just to show you that the total variability

 

      you are going to see in any test method is going to

 

      be the variability that is inherent to your product

 

      and your manufacturing process and the variability

 

      that is inherent to your test method.  For

 

      dissolution the variability inherent to the test

                                                                60

 

      method can be quite large, especially if you don't

 

      understand how all the different parameters can

 

      affect your product.

 

                I am going to just show some examples of

 

      some of the variability.  You can see I have a lot

 

      of information up on this slide, and every single

 

      one of these bullets can be a source of variability

 

      when running a dissolution test method.  You have

 

      to make sure your operators are well trained.  You

 

      have to make sure you have set things up properly.

 

      You have to make sure that you understand how all

 

      the different media and equipment parameters,

 

      sinkers etc., can affect the variability of your

 

      specific product.  So, there are a lot of places in

 

      here where, you know, if you add a tenth or so, or

 

      a percent or two of variability by the end you have

 

      quite a wide range of potential dissolution

 

      parameters you could get even with the same lot of

 

      material.

 

                When it comes to mechanical calibration, I

 

      think I showed some of the USP parameters earlier

 

      and what I want to show you here is actually that

                                                                61

 

      in our lab, DPA, we use more stringent mechanical

 

      calibration than what is listed in the USP.  A lot

 

      of the criteria we use come directly out of the

 

      PhARMA recommendation.  I think that paper is in

 

      your packet.  It came out in the '90s, where they

 

      did a collaborative study to take a look at

 

      mechanical calibration a little more closely to see

 

      if tighter mechanical calibration might reduce

 

      variability when running the calibrator tablet.

 

                Because we run so many products in our lab

 

      and we don't necessarily have the time to stop and

 

      see if this product is really sensitive to

 

      centering or not, etc., we just try to be very

 

      careful about how we set up our equipment.  Some

 

      tools are now available to very easily set these

 

      parameters much tighter than what is currently in

 

      the USP.  So, you can see that for quite a few of

 

      these we are tighter, and for others we have added

 

      criteria that are not actually in the USP as

 

      specific numbers.  For instance for shaft wobble

 

      and vibration, we actually measure those and set

 

      criteria for those.

 

                Degassing is one of the things I think

 

      that really got us into trouble--I don't want to

 

      use that word, but with the malaria drug the

                                                                62

 

      different labs were degassing in different ways and

 

      this drug happened to be very sensitive to

 

      degassing.  So, typically in the past the way you

 

      decided whether your media was well degassed or not

 

      is that you ran the calibrator tablet.  The 10 mg

 

      prednisone is very sensitive to dissolved gasses in

 

      the media so if you weren't sure if you were

 

      degassed or not you could just run that calibrator

 

      tablet to see if you were in range and then decide

 

      if you were degassed properly.

 

                Well, it turns out that there is some

 

      equipment on the market that you can use to

 

      actually measure dissolved gasses so this is

 

      something we have done recently in our lab.  We

 

      have taken this meter, which is actually used in

 

      other industries and not in the pharmaceutical

 

      industry, and used it to try to determine how much

 

      dissolved gases are left after using different

 

      degassing techniques.

 

                There are many different ways in which

 

      people degas their media.  The reason you need to

 

      degas your media is because there are some products

 

      that if you take a vessel and you drop in a tablet

 

      or capsule, what will happen is you have gases in

 

      the media.  The bubbles will form around this

                                                                63

 

      tablet or capsule and oftentimes will prevent it

 

      from dissoluting.  So, you actually need to get the

 

      gases out of there before you start.

 

                Here is a little graph of the different

 

      ways people degas and the results we got with the

 

      total gas meter, measuring both total gas and

 

      oxygen.  You can see that for the first bar over

 

      there that is obviously atmospheric pressure and

 

      atmospheric oxygen in the media.  These are all

 

      done in just plain water.  The next bar is the way

 

      we degas at DPA, which is point of vacuum at less

 

      than 150 ml of mercury with agitation, and you can

 

      see we get rid of about a little more than half of

 

      the total dissolved gases and quite a bit of the

 

      oxygen.

 

                The USP method is also very good.  There

                                                                64

 

      you are heating up to about 41 degrees and

 

      aspirating to remove the dissolved gases.  They

 

      also get half the total gone and about half the

 

      oxygen.

 

                Some people actually helium sparge and you

 

      can see helium sparging and although you do reduce

 

      the oxygen significantly you do not reduce the

 

      total dissolved gases.

 

                So, does this matter or not matter?  You

 

      know, this all depends on the product you are

 

      testing.  So, I just want to show you some examples

 

      here.  These are 3 different products, called

 

      product 1, 2 and 3 so I don't give any product

 

      names.  You can see that for product 1 and product

 

      2 there is a huge difference between non-degassing

 

      and degassing.  For both of those graphs I have

 

      shown two different ways of degassing.  One is the

 

      USP and DPA method, both of which give similar

 

      results.  The other is helium sparging.  You can

 

      see in both cases that the helium sparging does

 

      give slightly higher results than either the USP or

 

      the DPA method.  Certainly, for product 2 helium

                                                                65

 

      sparging gives much more variable results than the

 

      DPA degassing.  You can see that on the helium

 

      sparging line which is kind of the green-yellow

 

      one.

 

                You can see that product 3 doesn't really

 

      care whether you degas or not.  One of those lines

 

      is non-degassed and one is the DPA method which had

 

      the lowest percent of dissolved gases.  You can see

 

      that you get essentially very similar dissolution

 

      whether you degas or not.

 

                Larger than just degassing is the actual

 

      composition of the media.  I think as Ajaz

 

      mentioned, Japan is looking at what type of media

 

      you actually want to be using.  We see a lot of

 

      acid here and some buffers.  Here is a product and

 

      the dissolution method is pH 7.2.  So, 7.2, as you

 

      can see on your left I guess, is the media that is

 

      used in this product.  It also turns out that with

 

      these 6 different tablets there is some variability

 

      between the 6 but they all passed the dissolution

 

      specification for this particular product.

 

                This is a product where we wanted to take

                                                                66

 

      a look at some lower pHs just because there are

 

      some patients who happen to use this drug who may

 

      have lower intestinal pH than 7.2 and so we went

 

      down to 6.8 and, lo and behold, every single tablet

 

      looked different to us and no two tablets were the

 

      same.  We repeated this over and over again, trying

 

      to figure out what is going on.  You can actually

 

      do a lot with dissolution by just watching your

 

      product.  There is nothing like the human eye

 

      sometimes.

 

                If you watch this product in the vessel

 

      what you will see is that it sits there and does

 

      not dissolve and you get no dissolution until you

 

      see the coating split open.  Once the coating

 

      splits open, then it dissolves fairly quickly.  So,

 

      taking a look at that we were trying to figure out

 

      what could be the sources of variability of this

 

      product.  Is it the way we are handling it when we

 

      put it into the dissolution vessel?  Are we

 

      damaging the coating in some way?  Are these tablet

 

      differences real or is this the manufacturing

 

      process itself?  Do we have instrument variation? 

                                                                67

 

      These 6 tablets are in 6 different vessels so is

 

      there some difference in these vessels where maybe

 

      we have improper calibration or something?

 

                Well, after much investigation, what we

 

      found is that this is actually a product problem.

 

      If you cut open these tablets and take a look at

 

      the coating, not all of them have uniform coating.

 

      You can see there, on the left, one of the tablets

 

      that has a very uniform coating thickness.  Then

 

      every once in a while you ran across a tablet that

 

      had a void between the drug and the coating.  The

 

      drug is actually on the left side here; it is kind

 

      of the yellow sparkly stuff and the red is the

 

      coating.  So, some of the tablets had very uniform

 

      coating; some of the tablets had defects.  These

 

      defects were dissolving much faster or were

 

      breaking open, splitting open much faster than the

 

      ones that didn't have defects.  This is a situation

 

      where perhaps dissolution could help this

 

      manufacturer make a more consistent product if they

 

      were doing their dissolution at a slightly

 

      different pH or doing a dissolution test method at

                                                                68

 

      several different pHs to try to make sure they were

 

      making a consistent product.

 

                I was just going to mention sinkers

 

      because I talked about them and also because they

 

      do make a big difference.  The graph up there

 

      actually has nothing to do with the sinkers but it

 

      shows you what happens if you don't get your tablet

 

      at the center of your vessel.  The bottom blue line

 

      is product 1, right down at the bottom of the

 

      vessel, centered completely.  The green-yellow line

 

      is if it is off center by 1 cm.  So, if it is just

 

      off center by a centimeter you can see that it

 

      dissolves much faster.  There are different

 

      hydrodynamics in that area than at the bottom of

 

      the vessel.  So, if you have a tablet that is

 

      fairly light and is not going to stay put, then

 

      often you will put it inside a sinker.

 

                Traditionally, in our lab we have used the

 

      sinker at the top to the right.  That is the one

 

      that we have used in our lab.  It is very easy to

 

      use.  It has a spring load and you just pull back

 

      the spring and drop the capsule or tablet in and it

                                                                69

 

      is, you know, very convenient I guess.  The USP

 

      method is to use a wire and wrap the wire three

 

      times around the tablet or capsule.

 

                Well, we did run across a product--this is

 

      what I talked about, that you have to understand

 

      your product and how it reacts to different

 

      variables--that was sensitive to this actual

 

      commercial sinker.  This is the product we tested

 

      and with the commercial sinker that I just showed

 

      you it failed dissolution.  The specification here

 

      was 80 percent at 30 minutes and you can see that

 

      all 6 tablets failed.  Of course, we thought the

 

      product was perhaps a failure but it actually

 

      turned out that if you visually looked at what was

 

      going on, the product was being trapped.  It was

 

      swelling up and getting trapped inside that

 

      commercial sinker and so it could not essentially

 

      dissolve.

 

                We went back to the USP method with three

 

      wire turns around the tablet, and you can see that

 

      the product passes wonderfully with no problems

 

      whatsoever.  So, we no longer use commercial

                                                                70

 

      sinkers in our lab but a lot of people use them so

 

      I just wanted to make you aware of the fact that

 

      something as simple as a sinker can affect the

 

      individual product that you are looking at.

 

                So, what I have tried to show you is just

 

      some data that illustrates the fact that different

 

      products are sensitive to different parameters when

 

      you are doing dissolution, and there are obviously

 

      a lot of places where you can introduce variability

 

      in your test method.  What we would like to propose

 

      is an alternate approach to calibration and

 

      validation which includes complete understanding of

 

      how dissolution and the measurement system in your

 

      product specific variables affect variability, and

 

      try and understand the relationship between your

 

      product properties and your dissolution results.

 

      This includes understanding the dissolution

 

      apparatus that you are using, why you are choosing

 

      it and why you are choosing the media you are

 

      choosing, and determine, hopefully, the best method

 

      to give you opportunities for improvement and to

 

      ensure that the quality of your product is good.

 

                You can see that because of the way

 

      dissolution is currently set up there are a lot of

 

      things you have to control, and perhaps there are

                                                                71

 

      new approaches we can also use to get the same type

 

      of information that might have inherently less

 

      variability.  Then, obviously, a part of this whole

 

      process needs to be communication and training.  If

 

      people are out there saying that FDA wants us to

 

      use apparatus 1 and 2, then that is what people are

 

      going to do.  So, the FDA is trained in a more

 

      open-minded look at other things.  If people feel

 

      that way at least, then they might be willing to

 

      look at other approaches.

 

                When it comes to alternative approaches to

 

      dissolution calibration validation, I think as I

 

      told you in our lab we do more stringent mechanical

 

      calibration because some products are very

 

      sensitive to how the apparatus is set up and,

 

      certainly, if you set it up properly your

 

      variability will be less than the variability of

 

      the calibrator tablet.  Certainly, when you are

 

      using your specific product itself, you need to ID

                                                                72

 

      and control all the source of variability that you

 

      are going to see.  You need to determine how your

 

      product is sensitive to things like the apparatus

 

      type, the setup parameters and the media, both type

 

      of media and whether it is degassed or not.  There

 

      is an interaction between the instrument you use

 

      and your product, and understanding that is going

 

      to also help you reduce the variability in the

 

      dissolution test method.  People like to use

 

      calibrator tablets.  I think it gives them a

 

      measure of confidence that they set everything up

 

      and their system is suitable.

 

                So, what we are proposing is that

 

      certainly the USP calibrator can be used if

 

      somebody wants to take a look and see that they

 

      have set up properly.  Perhaps it also might be

 

      useful to set up an internal calibrator maybe based

 

      on a bio. batch or clinical batch to make sure of

 

      system suitability.  The calibrators dissolve in a

 

      certain way or are sensitive to certain things and

 

      not sensitive to certain things, the USP ones, and

 

      those parameters may not be the parameters that

                                                                73

 

      your particular product is or is not sensitive to.

 

      So, creating your own internal calibrator and

 

      understanding how your product is sensitive to all

 

      the parameters is going to be perhaps better than

 

      an outside product that may not have the same

 

      sensitivities that yours does.  Obviously, you need

 

      to confirm the suitability of your internal

 

      calibrator using some kind of a gauge R&R study so

 

      you can really understand what the variability is

 

      in your product.

 

                Ajaz mentioned gauge R&R a little bit.  If

 

      you pick a lot of product or a piece of a lot to

 

      maybe set up as an internal calibrator you need to

 

      carefully characterize that and determine what its

 

      variability is.  You want to make sure it is

 

      representative of your manufacturing process.  You

 

      want to make sure that it was manufactured while

 

      your process was under control.  Obviously, when

 

      you are doing a gauge R&R you need to take a look

 

      at what variability is introduced instrument to

 

      instrument, vessel to vessel.  As you can see, each

 

      instrument is like 6 individual little instruments.

                                                                74

 

      And variability from personnel to personnel and,

 

      obviously, media and whether it is degassed or not.

 

                We need to understand the benefits and

 

      limitations of the different dissolution apparatus.

 

      I showed you that there are 7 different ones in the

 

      USP.  We also sometimes get ones that are non-USP

 

      apparatus when people submit test methods.  So,

 

      there are a lot of different things out there to

 

      choose from and, better than just choosing one that

 

      someone thinks maybe the FDA wants to see, maybe

 

      try to understand how the hydrodynamics work; try

 

      to model your system.  Actually, I have been told

 

      by people who do modeling that apparatus 1 and 2

 

      are difficult to model so there may be some better

 

      systems out there where we can do some better

 

      predicting of what is going to happen as we change

 

      physical parameters of our product, and take a look

 

      at some other things we might be able to do.

 

                Of course, what would even be better is

 

      just quit doing dissolution as it is known today

 

      and maybe find some other ways to assess product

 

      quality.  People have done some work in our sister

                                                                75

 

      lab here, in White Oak, to try to correlate

 

      dissolution with NIR.  There is a lot of

 

      spectroscopy out there that can be used online as

 

      part of a PAT feedback loop, and perhaps good

 

      correlations and good models could be developed

 

      between those and quality and in vivo availability

 

      and we can dispense perhaps with the current

 

      dissolution test method, which has all of its

 

      parameters--things that can go wrong and need to be

 

      set very carefully.  Obviously, key to this is

 

      going back to the first principles and modeling and

 

      understanding your formulation, and how each

 

      component of your formulation contributes to the

 

      quality of your product.

 

                So, that is all I had to say and I just

 

      wanted to acknowledge Terry Moore, who is actually

 

      here today, who probably knows more about

 

      dissolution than anybody in the world.  He is

 

      sitting over there, if you want to know more about

 

      dissolution.  Then, Zongming Gao is also in our lab

 

      doing dissolution; and Lawrence who also knows a

 

      lot about dissolution; and Ajaz all helped with

                                                                76

 

      this.  So, thank you.

 

                DR. COONEY:  Thank you very much, Cindy.

 

      There certainly is time for questions.  Gerry?

 

                     Questions by Committee Members

 

                MR. MIGLIACCIO:  Cindy, first I applaud

 

      your last comment about using alternate methods.  I

 

      just want to point out that you made several

 

      comments about the use of apparatus 1 and 2 and,

 

      speaking I think for most companies, we don't use 1

 

      and 2 because we think FDA wants us to use them.

 

      You did a great job of pointing out the variability

 

      of the different parameters that can impact

 

      variability.  It is very important when you are

 

      testing thousands of batches a year that you have a

 

      really well trained work force that knows how to

 

      use this apparatus, and that you have consistency

 

      in the way you test because if you are switching

 

      from one apparatus to another it presents another

 

      level of complexity.  So, it is really the

 

      consistency.  Because of the variability that is

 

      inherent here, it is the consistency that drives us

 

      to apparatus 1 and 2 and not a lack of desire--

 

                DR. BUHSE:  To try something else?

 

                MR. MIGLIACCIO:  --but, you know, it is

 

      complicated enough so it is really consistency that

                                                                77

 

      drives us there.

 

                DR. COONEY:  Marvin?

 

                DR. MEYER:  The data you showed from your

 

      lab versus the specs on prednisone, and you said in

 

      one case you tend to be high and some cases fail,

 

      when you do fail the calibration what do you do

 

      about it?  Is it the calibration that is no good?

 

      Is it the USP specs that is no good?  Is it the lab

 

      that is no good?  Or, do you just keep going until

 

      you have 36 samples?

 

                DR. BUHSE:  Well, historically what we

 

      have done is double check your mechanical

 

      calibration and then you really run the calibrator

 

      tablet.  So, was the original failure the tablet?

 

      Rarely do we find something to adjust when we check

 

      the mechanical calibration.  We do the mechanism

 

      calibration much tighter than the USP anyway so

 

      essentially you rerun.  We actually don't run them

 

      anymore in the lab, the USP calibrator tablets.

 

                DR. MEYER:  That solves that problem!

 

                DR. BUHSE:  That solves that problem!  We

 

      have an internal calibration tablet that we use now

 

      that we have characterized ourselves in our lab

 

      that has lower variability.  We stopped using this

 

      one probably at the end of last year.  The data I

                                                                78

 

      showed was the data from 2004, 2003.

 

                DR. MEYER:  The other question I have or

 

      comment is that on one of the slides you suggest

 

      using perhaps an internal calibrator, a bio. batch

 

      or some known that you have produced.

 

                DR. BUHSE:  Right.

 

                DR. MEYER:  How do you know that that

 

      product, over the lifetime of the product being

 

      manufactured, hasn't changed?  Dissolution doesn't

 

      change, you are satisfied your equipment is in good

 

      order when, in fact, it isn't because you couldn't

 

      pick up the change--

 

                DR. BUHSE:  Stability is a big issue.

 

      Stability is an issue with the current USP

 

      calibrator.  It is known to drift down I believe

 

      with the paddle method over time, or whatever.  Do,

                                                                79

 

      you want to talk about that, Ajaz?

 

                DR. HUSSAIN:  Yes.  Marvin, I am going to

 

      go over that in detail.  The gauge R&R is actually

 

      for three purposes.  It is to establish and

 

      benchmark the variability.  I think the proposal

 

      actually is that mechanism calibration actually is

 

      sufficient.  The gauge R&R is an opportunity to

 

      establish your target.  You benchmark your

 

      variability and then use that variability for

 

      setting specifications, and so forth.  But then you

 

      have that and then you can keep the system stable.

 

      I think stability of the system has to be based on

 

      mechanism calibration.  That is what other

 

      countries do anyway.  So, I will go over that in a

 

      bit more detail.  So, the opportunity is more than

 

      just the internal calibrator.  So.

 

                DR. MEYER:  One follow-up, I kind of joked

 

      that you made the problem go away because you are

 

      not using it anymore.  What if you are a company

 

      and had in your NDA or ANDA that you would

 

      calibrate your dissolution using the prednisone and

 

      USP and you started to fail, your dissolution

                                                                80

 

      couldn't meet the calibration?  They don't have the

 

      luxury of just saying, well, we are going to use

 

      our own now because they are stuck with using what

 

      they said in the NDA, right?  What should a company

 

      do about that?

 

                DR. BUHSE:  You want to talk about that,

 

      Ajaz?

 

                DR. HUSSAIN:  Well, I think this meeting

 

      is step one to start addressing that in a sense.

 

      Here is an alternate procedure.  So, I think if the

 

      advisory committee will sort of endorse this and we

 

      move that way, we will put that in policy and there

 

      are many different ways to implement that.  So.

 

      But from the compendia perspective, I think you

 

      have to comply with the compendia so that is a

 

      different challenge that the industry and companies

 

      have to deal with.  So, all we are doing right now

 

      is creating an alternate regulatory decision

 

      pathway and our enforcement strategy based on that.

 

                DR. COONEY:  Nozer?

 

                DR. SINGPURWALLA:  Slide number 13, I

 

      thought you said it was Ajaz's slide.  Therefore,

                                                                81

 

      it is wrong!

 

                [Laughter]

 

                DR. BUHSE:  Yes, it was Ajaz's slide.

 

                DR. SINGPURWALLA:  Well, how do you

 

      distinguish between repeatability and

 

      reproducibility?

 

                DR. BUHSE:  Well, I was going to say with

 

      a destructive test it is very difficult.

 

                DR. HUSSAIN:  See, this is gauge R&R for a

 

      destructive test.  You really have to have design

 

      experiment and I was going to cover that in my

 

      talk.  What this does is, it actually ensures that

 

      the lot you choose is stable and in a state of

 

      control.  That is the only way you can actually

 

      move in this direction.  So, that achieves that

 

      target.  The destructive gauge R&R is a very formal

 

      experiment and it is a nested design which does get

 

      an estimate of whether a practice or an operator

 

      can repeat it.  That is repeatability.

 

      Reproducibility is the variability associated with

 

      that.

 

                DR. SINGPURWALLA:  So, the repeatability

                                                                82

 

      refers to a physical thing.  The other thing is I

 

      don't know how important it is for you to manage

 

      variability but if it is important to you to manage

 

      variability, then my sense is that as the product

 

      variability increases the measurement variability

 

      will also increase.  Therefore, there will be

 

      correlation and, therefore, the sigma squared total

 

      that you have will be underestimated the way you

 

      have put it down.  If it is of any importance, you

 

      may want--

 

                DR. HUSSAIN:  I think it is.  That is the

 

      reason the leverage--the quality by design having

 

      the pharmaceutical development information starts

 

      to allow us to dilute some of this.  But the

 

      variability that you are observing you are

 

      observing to the eyes of the measurement system so

 

      the measurement system and variability in the

 

      product are together.  I will try to come back and

 

      sort of explain some of that.

 

                DR. DELUCA:  I apologize for my voice.

 

      You very nicely pointed out the multitude of

 

      variables that are involved.  There is instrument

                                                                83

 

      variability as well as product variability so you

 

      have interaction.  You mentioned degassing.  But

 

      you are using a set agitation in your system.  When

 

      you start degassing, are you not sparging?  Now,

 

      you can create agitation or sparging during the

 

      test?

 

                DR. BUHSE:  No, it is done beforehand.

 

      You do it before you start and you put the media in

 

      the different vessels and there is no degassing

 

      during dissolution itself.  Questions come up,

 

      especially for extended-release products, where

 

      actually the dissolution test method lasts for 24

 

      hours per product, and the question then becomes

 

      what happens to the gas level over that time.  We

 

      hope to test that with this meter.  The one I

 

      showed you here is actually one that has a probe

 

      that is, like, 3 inches around so you have to put

 

      it in a giant vessel.  They are making a new probe

 

      that is small and will fit inside the dissolution

 

      vessel so we can see what happens actually in the

 

      dissolution vessel over time.  Like you say, with

 

      some of these test methods at high rpm, 100 rpm, we

                                                                84

 

      are getting a lot of agitation.  So, that is a good

 

      question.

 

                DR. DELUCA:  And I was worried about the

 

      product and how product variation can affect--so,

 

      you have an interaction between the instrument and

 

      the product where particle size might influence,

 

      you might have a set agitation rate.  If the

 

      particle size changes then it is going to change

 

      the result.

 

                DR. BUHSE:  Right, unless you have a

 

      method that can discriminate that if it is

 

      important to the acting of the drug.

 

                DR. DELUCA:  You have talked about

 

      modeling, I mean you mentioned it.  Maybe it is

 

      going to be covered later on, but I wondered if you

 

      include anything here to look at profiles, release

 

      profiles.

 

                DR. BUHSE:  We haven't done a lot of

 

      modeling yet in our lab.  I don't know if we are

 

      going to talk about that specifically later on or

 

      not today.

 

                DR. COONEY:  Ken?

 

                DR. MORRIS:  Just a couple of things.  One

 

      is that given the sort of lag--I guess I just have

 

      a philosophical problem with calibrator tablets in

                                                                85

 

      that if you are looking at a process and want to

 

      independently establish that it is in control or

 

      that it is doing what you think it is doing--we are

 

      producing these the same way we produce the tablets

 

      for testing--

 

                DR. BUHSE:  That are no better.

 

                DR. MORRIS:  What is that?

 

                DR. BUHSE:  That are no better.

 

                DR. MORRIS:  In fact, there are some data

 

      that I think we will see to day that there are some

 

      liabilities.  I think maybe this is something we

 

      will talk a lot more about, I am sure, but I think

 

      one of the things that may come out of this is that

 

      calibrator tablets just don't have a prominent

 

      role.  What I would say is that if you look at an

 

      immediate-release system--and we will also get into

 

      BCS exemptions--then the issues become sort of

 

      treatable in other ways.  If you are looking at

 

      sustained-release or modified-release, such as

                                                                86

 

      enteric or extended, then my argument is that you

 

      ought to be controlling the coating process and

 

      that sort of activity is really much more advanced

 

      than it was.  I mean, you have your example of the

 

      tablet that has the air pocket but probably what

 

      was more important was the difference between the

 

      80 and 50 micron coating thickness.  This is

 

      clearly a failure of reproducibility of coating and

 

      the dissolution may catch it or may not.  I mean,

 

      the statisticians--I don't know, there is the

 

      Bayesian argument but I have talked to Sandy

 

      Bolton, for one so, you know, if you have high

 

      variability dissolution maybe 6 tablets is enough

 

      to pick it up but, depending on what constitutes

 

      high variability, you know, it is in the laps of

 

      the gods whether you get it or not.  So, to the

 

      extent that things are surface-based alternate

 

      methods--I mean, in the first place, you want to be

 

      controlling the coating processes and then, to the

 

      extent they are surface-based, have you considered

 

      things within the group like the combination of

 

      that and, like, IGC to look at surface free

                                                                87

 

      energies or something that is at least a little

 

      less subjective?  I don't know if you have talked

 

      about it because everything else is a correlated

 

      technique.

 

                DR. BUHSE:  Right.

 

                DR. DELUCA:  Whereas, something that

 

      actually measures surface free energy, even though

 

      there is no practical instrument right now, is a

 

      direct measure.

 

                DR. BUHSE:  We haven't done that with that

 

      particular product.  We have tried to do some

 

      spectroscopy correlations.

 

                DR. HUSSAIN:  If I may?

 

                DR. COONEY:  Yes.

 

                DR. HUSSAIN:  I think you make a good

 

      point, and I think the goal that we have, number

 

      two, desired state, specification based on

 

      mechanistic understanding--so, if the mechanism is

 

      controlling the dissolution based on a coating

 

      thickness, if you are able to measure the coating

 

      thickness reliably, and so forth, that should be

 

      sufficient.  So, I think that is the direction we

                                                                88

 

      wish to move in, and some of the new technologies

 

      and science sort of helps that.

 

                There is another point I think which I do

 

      want to make and this is my graduate school

 

      training; this is biopharmaceutics 101.  When we

 

      approach trying to develop a product we first think

 

      about the patient, and so forth.  Prof. Richard

 

      always insisted you don't even think about an in

 

      vitro test.  You first try to get initial

 

      information in humans and then say, all right, what

 

      sort of testing will we need.  So, you establish

 

      your formulation, human connection or patient

 

      connection first before spending time in an

 

      artificial way.  In my consulting role before I

 

      came to FDA, one company I worked for carried out

 

      53 experiments, screening and so forth; they had no

 

      idea whether dissolution was useful or not.  They

 

      spent all this development effort trying to

 

      optimize a hypothetical, what they thought was the

 

      dissolution rate and the first experiment they did

 

      was completely off.  So, all the experiments were

 

      actually off target.

 

                So, there is a tendency within industry to

 

      assume that in vitro dissolution is going to guide

 

      them to a formulation without even understanding

                                                                89

 

      its relevance.  I think Marvin knows that company

 

      very well.  We actually had a paper on that issue

 

      together.  So, there are challenges I think.  So,

 

      quality by design actually forces us to think what

 

      is the patient and then think about the tests so

 

      that is what we are trying to achieve here.

 

                DR. COONEY:  Ajaz, perhaps we can capture

 

      that as a point, that the purpose of formulation

 

      development is to optimize patient care, not

 

      dissolution assay.  We hear you.

 

                DR. FACKLER:  Could I just make a point?

 

      Dissolution can function for a number of different

 

      purposes and on one of your slides you suggested

 

      that finding a discriminating method might be

 

      useful, and I would agree under certain

 

      circumstances.

 

                On the other hand, if you look at that

 

      enteric-coated product really the purpose of the

 

      enteric coating is to protect the tablet for the

                                                                90

 

      first hour, or whatever time it might exist at the

 

      very acidic condition of the stomach.  Whether or

 

      not coating breaks open at one hour, two hours or

 

      three hours might have no relevance to the in vivo

 

      performance of the product.

 

                So, I think it is important, as we talk

 

      about the future of dissolution testing, to

 

      recognize what it is intended for.   If it is

 

      intended to predict in vivo performance, that is

 

      one thing and a predictive or correlative method

 

      then I think would be the ideal.  If it is to look

 

      for product quality and to reduce the inherent

 

      variability in products, well, then a more

 

      discriminating method that might have no relevance

 

      to in vivo performance might be our goal.  I think

 

      we just need to keep that in perspective as we

 

      think about the future of dissolution testing.

 

                DR. HUSSAIN:  If I may since we have time,

 

      I think this is one of the first steps in our

 

      tactical plan.  Since we have time, if we could

 

      engage the advisory committee to make sure is this

 

      an acceptable step further discussion is needed. 

                                                                91

 

      So.

 

                DR. COONEY:  Tom?

 

                DR. LAYLOFF:  Yes, I was going to say

 

      because of my concern with the problem with

 

      degassing--I never degas my stomach before I take

 

      my medication--

 

                [Laughter]

 

                DR. FACKLER:  You probably don't swallow

 

      900 ml of water either.

 

                [Laughter]

 

                DR. MORRIS:  Just a couple of comments.

 

      First, when we validate equipment we have to

 

      understand what tests we are doing and what we are

 

      trying to validate, and the standard tablet just

 

      doesn't--intuitively, it doesn't get there for me

 

      because we are looking at validating a piece of

 

      equipment and all of a sudden the variables that we

 

      are throwing into the pot include what is the

 

      dissolution medium and how we handle that; what is

 

      the size of tablet and how we handle that when we

 

      are trying to validate a piece of equipment.  So,

 

      probably the first step is saying what validates

                                                                92

 

      the equipment, and anything else we do is a waste

 

      of time.

 

                Then, the next step, to get right to what

 

      Paul said, is what is my dissolution test telling

 

      me because I am a manufacturer and I want to keep

 

      my process under control, or am I predicting what

 

      is happening in people?  We have seen for 35 years,

 

      as far as I know, that dissolution doesn't predict

 

      the human results in terms of bioavailability or

 

      bioequivalency.  You can't do it that way.  You

 

      have to get that data and then try to correlate.

 

      So, if we are using dissolution for quality

 

      control, for process, fine, then there is a set of

 

      variables and we do it that way.  But if we are

 

      trying to say that I can do a dissolution study

 

      and, therefore, I will know that my formulation is

 

      going to work in a person I think we are really

 

      biting way more off than we can chew.

 

                DR. HUSSAIN:  I think I agree with you,

 

      but in may aspects you do establish correlation.

 

      Actually, Lawrence, in his talk, will actually make

 

      that same proposal as you did.  So.

 

                DR. COONEY:  Tom?

 

                DR. LAYLOFF:  The early work done on

 

      digoxin was designed to go for in vivo/in vitro

                                                                93

 

      correlation for about 35 manufacturers, and that is

 

      how that standard was set.  Prednisone subsequently

 

      was done the same way.  In reviewing that, it would

 

      determine that if the FDA continued down that path

 

      it would eventually take all the resources of the

 

      FDA to do it because of the cost of performing

 

      those in vivo/in vitro correlations.  Then the

 

      dissolution standard was just arbitrarily applied

 

      across the board.

 

                DR. COONEY:  Marvin?

 

                DR. MEYER:  Ajaz, I think you ask if you

 

      are on the right track and I think you definitely

 

      are.  You know, when you first said we are going to

 

      revisit dissolution I said, oh, my God--

 

                [Laughter]

 

                --so, I think you are on the right track.

 

      I mean, for me, when I used to do some dissolution

 

      just in a university laboratory, I loved the wide

 

      range for the calibrators because then my equipment

                                                                94

 

      always passed and I didn't have to worry about it.

 

      But now, sitting around this table, I have a

 

      different hat on and it is shocking, 51-81 percent.

 

      How can you have a calibrator--if somebody comes in

 

      with analytical data like that you would say go

 

      away; this is a very poorly controlled analytical

 

      procedure.  So, I think that revisiting the issue

 

      is very important.

 

                DR. HUSSAIN:  Marv, in may ways, you know,

 

      I was blind to this.  I actually was not fully

 

      aware of the scenario, and Cindy will attest to

 

      this.  When I started writing this paper I put

 

      Lawrence through hell.  I said how could this

 

      happen?  Because our standard criteria for

 

      specification is plus/minus 10 percent and the

 

      instrument is this way so there was a disconnect

 

      that I was not aware of and I have to apologize for

 

      that.

 

                DR. COONEY:  Are there any other comments

 

      or questions at this point?

 

                [No response]

 

                What I would like to suggest is that we

                                                                95

 

      take a break for 15 minutes and reconvene at 10:25,

 

      and we are in good shape for continued discussion

 

      and I have no doubt there will continue to be more.

 

                [Brief recess]

 

                DR. COONEY:  I would like to now welcome

 

      Dr. Mehta to speak to us about an overview of the

 

      current guidance on the documents and decision

 

      process in biopharmaceutics.

 

              Overview of Guidance Documents and Decision

 

                   Process:  Biopharmaceutics Section

 

                DR. MEHTA:  Good morning.  As you can see

 

      on my slide here, I am asked to give an overview of

 

      guidances documents and decision processes from a

 

      biopharmaceutics perspective.

 

                Before I start, I want to acknowledge a

 

      couple of people in my division, Dr. Ramana Uppoor,

 

      she is sitting in the back in the audience, and Dr.

 

      Patrick Marroum, team leaders in neuro. and

 

      cardiorenal in my division and some of the experts

 

      in biopharmaceutics in my division.

 

                This is the outline of my presentation.  I

 

      am going to give you an overview of

                                                                96

 

      biopharmaceutical aspects of dissolution-related

 

      guidances.  That is a formidable task.  My first

 

      draft that I sent to Ajaz had 100 slides and Ajaz

 

      replied by saying an excellent overview but cut it

 

      down.  So, I am now down to 60.

 

                [Laughter]

 

                But I still intend to finish in time.

 

      Then with a quick overview I will take you through

 

      some examples from our NDA reviews of

 

      immediate-release and modified-release products,

 

      and share with you my perspective on opportunities

 

      for improvement.

 

                These are the guidances I am going to

 

      quickly take you through.  Chronologically they are

 

      different but in terms of science, the way the

 

      ideas are represented I have shifted them around.

 

      I am going to first start with the BCS guidance.

 

      In parentheses are the references.  I will follow

 

      that by the immediate-release dissolution guidance

 

      that came out in 1997.  The BCS guidance was

 

      finalized in 2000.  The IR dissolution guidance

 

      invokes BCS principles and that is why I have

                                                                97

 

      arranged it that way.  That will be followed by a

 

      quick overview of the IVIVC guidance and that is

 

      for modified-release products, in vitro/in vivo

 

      correlation.  Then a couple of slides on general

 

      bioavailability and bioequivalence guidance, which

 

      was finalized in 2003.

 

                I will quickly switch to something known

 

      as scale-up and post-approval changes for

 

      immediate-release products and modified-release

 

      products, and the topics covered there.

 

                So, let me start with the BCS guidance

 

      summary.  Maybe it is known to everybody, but just

 

      for the sake of completeness let me point out the

 

      highlights of the BCS guidance.  This guidance

 

      takes into account three major factors that govern

 

      the rate and extent of drug absorption from the

 

      immediate-release solid oral dosage form.

 

                These are the solubility and intestinal

 

      permeability of the drug substance, and dissolution

 

      of the drug product.  So, based on the solubility

 

      and permeability characteristics of the drug

 

      substance the drugs are classified into four

                                                                98

 

      categories: high solubility, high permeability; low

 

      solubility, high permeability; high solubility, low

 

      permeability; and then the fourth category, low

 

      solubility, low permeability.

 

                The third bullet is the central idea, the

 

      central concept, a very sound scientific concept of

 

      BCS which is, you know, if a drug product is BCS

 

      class 1, and for different formulations of this

 

      class 1 product if they are rapid and similarly

 

      dissolving you can give a biowaiver for the test

 

      formulation without requiring an in vivo

 

      bioequivalency assessment, provided you show

 

      similar dissolution profiles over the physiological

 

      pH range.

 

                The last important point about this

 

      guidance is that in this guidance we have defined

 

      what determines rapid dissolution, and we say if

 

      your drug product dissolves 85 percent in 30

 

      minutes over the pH range absorption should not be

 

      dissolution limited.  So, that is all for BCS.

 

                Moving on quickly to the immediate-release

 

      dissolution guidance summary, and again I will do

                                                                99

 

      my little bit of acknowledgement here, Dr. Shah and

 

      some members on the panel here have contributed to

 

      this guidance and, from my personal perspective,

 

      this is scientifically a very well written document

 

      although it was almost ten years ago.

 

                These are the topics covered in this

 

      guidance.  The guidance lays out approaches for

 

      setting dissolution specifications for a new

 

      chemical entity.  As I said, it takes into

 

      consideration BCS nature of the drug product and,

 

      depending upon that, you can have minimal

 

      dissolution requirements in setting specifications

 

      or more stringent.

 

                Another very important point from my

 

      perspective is that this guidance has outlined

 

      something known as mapping or response surface

 

      methodology.  Again, this is supposed to be for

 

      immediate-release products.  The guidance says that

 

      undefined clinical manufacturing

 

      variables--manufacture your products at the

 

      extremes of CMVs and in vivo performance and, if

 

      you have that information, you will have a very

                                                               100

 

      sound rationale for coming in with appropriate

 

      dissolution specifications.

 

                Finally, in this guidance there is a

 

      discussion of how do you compare dissolution

 

      profiles of two products.  One of the approaches

 

      that I recommend is known as the f2 or the

 

      similarity factor which essentially looks at the

 

      differences in dissolution at each time point, with

 

      a range of 0-100.  An f2 of 50 or greater than 50

 

      indicates similarity of the dissolution profiles.

 

      As we have said in that guidance, dissolution

 

      specifications are established in consultation with

 

      Biopharmaceutics and the CMC review staff. The

 

      general bioavailability/bioequivalence guidance

 

      summary, again limited only to dissolution

 

      considerations, we have a section in there that

 

      talks about what should be submitted in an NDA or

 

      an ANDA in terms of a dissolution method.  There

 

      should be a dissolution method development report

 

      for an NDA, new drug application.  It should

 

      contain a pH solubility profile of the drug

 

      substance; dissolution profiles generated at

                                                               101

 

      different agitation speeds; and dissolution

 

      profiles generated on all strength in at least

 

      three dissolution media.  Essentially you want to

 

      see the in vitro performance of your product over a

 

      variety of conditions, including different media

 

      and different agitation; and select the agitation

 

      speed and medium that provides adequate

 

      discriminating ability, taking into account all the

 

      available in vitro and in vivo data.

 

                For ANDAs, abbreviated new drug

 

      applications, the guidance states that one should

 

      start with an appropriate USP method if it is

 

      there, in the USP.  For some reason, if it is not

 

      there for this product, then if the FDA method is

 

      publicly available, utilize that.  If that is not

 

      available, also publicly available, then submit the

 

      dissolution method development report, as described

 

      above for a new drug application.

 

                Again, for modified-release products for

 

      ANDAs the dissolution profiles use the appropriate

 

      USP method, if available, otherwise use the FDA

 

      method for the reference listed drug if available. 

                                                               102

 

      In addition, and I think this is probably because

 

      you could have for a generic similar or different

 

      release mechanisms, so additional dissolution data

 

      in three different media.

 

                Now switching to the IVIVC guidance which

 

      is, you know, in vivo/in vitro correlation for

 

      modified-release products, again from my

 

      perspective, this is a very useful guidance also.

 

      The main purpose of this guidance was to provide an

 

      outline for waiver of bioequivalency studies for

 

      modified-release products if one was able to

 

      establish an in vivo/in vitro correlation, a

 

      quantitative correlation.

 

                The guidance defines correlation in

 

      different categories, A, B, C and D.  Level A

 

      correlation is most quantitative, and I have listed

 

      in my presentation just the level A discussion.

 

      Level A correlation is supposed to be a

 

      point-to-point relationship between the in vitro

 

      dissolution and the in vivo input rate of the drug

 

      from the dosage form.  Usually this is a two-stage

 

      process, meaning that you take your dissolution

                                                               103

 

      data, convert that into dissolution rate, and you

 

      take your in vivo data and convert that into

 

      absorption rate and correlate the two.  Generally,

 

      this relationship is linear but non-linear

 

      relationship is also acceptable provided it is

 

      adequately characterized.

 

                So, this is an example of how level A

 

      IVIVC would look.  On the Y axis you have percent

 

      of drug absorbed and on the X axis is the percent

 

      of drug dissolved; your linear relationship over

 

      the range and this establishes your correlation.

 

      For the purpose of obtaining biowaivers, you need

 

      validation of this level A correlation.  From the

 

      point of view of setting dissolution

 

      specifications, that level of validation is not

 

      necessary, and I will get into that subsequently in

 

      my examples.

 

                In the IVIVC guidance for modified-release

 

      products we have some general concepts laid out for

 

      what the dissolution specification should mean.

 

      Ideally, as we say in the guidance, all lots within

 

      the lower and upper limit of the specifications

                                                               104

 

      should be bioequivalent.  At the minimum, those

 

      lots should be bioequivalent to the clinical trials

 

      lots or an appropriate reference standard chosen by

 

      the agency.  In other words, you have your

 

      reference performance and the upper limit should be

 

      similar to the reference and the lower limit should

 

      be similar to the reference.  Ideally, the extremes

 

      should be bioequivalent.

 

                Some further considerations are that

 

      variability alone should no longer be a primary

 

      consideration in setting specifications for

 

      modified-release products.  Specifications wider

 

      than 20 percent are acceptable only when evidence

 

      is submitted that lots with mean dissolution

 

      profiles that are allowed by the upper and lower

 

      limits are bioequivalent.  In other words, you can

 

      have specifications wider than 20 percent if you

 

      have a correlation, a quantitative correlation.

 

                If you don't have an IVIVC and you want to

 

      set dissolution specifications for modified-release

 

      products, these are some of the characteristics of

 

      what the data should be.  The profile should have

                                                               105

 

      at least three time points.  The last time point

 

      should be the time where 80 percent of the claimed

 

      labeled amount is dissolved.  Specifications are

 

      set to pass at stage 2, meaning that there are 12

 

      dosage units.

 

                As I mentioned a while ago, for setting

 

      dissolution specifications with the IVIVC, external

 

      validation is not required and, as I already

 

      mentioned, wider specifications based on what the

 

      correlation predicts can be done.

 

                This is graphically presenting that.  On

 

      the left panel you see that in the middle is the

 

      performance of your product, the variability around

 

      the mean dissolution profiles.  The blue line is

 

      the upper limit of the specification.  The red line

 

      or orange line is the lower limit of the

 

      specification.  You take that data using your in

 

      vitro/in vivo correlation model.  You predict the

 

      plasma concentration based on the two limits.

 

                On the right panel, the diamonds are the

 

      actual blood levels, the predicted blood levels at

 

      upper and lower limit, and the predicted level for

                                                               106

 

      Cmax and AUC should not be greater than 20 percent.

 

      Back in '97, what we could come up with was setting

 

      the consideration based on the mean difference.

 

      So, the upper and lower limit would not differ on

 

      the mean AUC and Cmax by 20 percent.  We could not

 

      build into this consideration the variability

 

      aspects and, as we have already heard in an earlier

 

      presentation today, that is an opportunity for

 

      improvement for future consideration.

 

                Switching gears, I am going to quickly

 

      tell you about what the SUPAC guidances mean as far

 

      as immediate-release and modified-release products.

 

      There are also a few guidances that came out

 

      subsequent to the issuance of the SUPAC in 1997,

 

      which is called equipment addendum, FDAMA and the

 

      changes approved to an NDA or ANDA guidance in

 

      2000.  Again, I am going to try to capture this

 

      very quickly.

 

                Conceptually speaking, these guidances

 

      identify what are the changes or what are the

 

      variables that are covered in terms of

 

      manufacturing considerations.  The level of changes

                                                               107

 

      for these variables, what are they?  They are

 

      defined; and then how do you deal with that?

 

                So, the variables covered in this

 

      guidance, manufacturing related, are composition

 

      and components.  For excipients it is

 

      non-release-controlling as well as

 

      release-controlling.  The non-release-controlling

 

      aspect is what is the part of the SUPAC-IR

 

      guidance.  That is taken as it is into the SUPAC-MR

 

      guidance and then what is added is the

 

      considerations for release-controlling excipients.

 

      Other variables covered are site, batch size,

 

      meaning scale-up and scale-down, manufacturing

 

      equipment and manufacturing process.

 

                I am going to take you through only one

 

      set of variables here and show you how the levels

 

      are defined and what are the related tests

 

      recommended and what are the related filing

 

      requirements.

 

                Essentially, the idea is this, the

 

      guidance has defined the level of change into three

 

      categories, level 1 is the minor change; level 2 is

                                                               108

 

      the moderate change; and level 3 is the major

 

      change.  So, moderate could have an in vivo impact

 

      on level 3 or major changes likely to have an in

 

      vivo effect.

 

                Related to those changes, the tests go

 

      along with them in terms of document evidence.  The

 

      lowest level, level 1, would usually require only

 

      application of compendia tests and stability data.

 

      Level 2 change would require extensive in vitro

 

      dissolution and release data.  That typically means

 

      that for immediate-release products you require

 

      profile comparison in five different media.  Then,

 

      for modified-release you need profile comparison in

 

      three different media.  Level 3 is the most

 

      significant change and that will be allowed only if

 

      you have an in vivo bioequivalency study or you had

 

      established in vitro/in vivo correlation.

 

                The filing requirements, again going from

 

      minimal to most which is annual report, changes

 

      being effected supplement, or prior approval

 

      supplement.  In the subsequent discussion I will

 

      just focus on the first two bullets, which is level

                                                               109

 

      of change and the tests.  I am not going to touch

 

      filing documentation at all.

 

                Here is an example of how the guidances

 

      break down changes into different levels.  For

 

      SUPAC-IR excipient levels excipients are listed for

 

      level 1 change, level 2 and level 3.  If you look

 

      at glidant, for example, for talc, plus/minus one

 

      percent change is allowed.  If you look at the top

 

      of the right-hand column, it is percent change

 

      weight of the change of the excipient over the

 

      weight of the total unit.  For talc it is

 

      plus/minus one percent.  Other glidants would be

 

      plus/minus 0.1 percent.  So, that is the lower

 

      limit of change, plus/minus 0.1 for talc.  If you

 

      look at filler, for example, it is also plus/minus

 

      five percent change.  So, this defines level 1

 

      change, minimal change.

 

                If you go to level 2 the ranges double.

 

      So, you go from plus/minus 0.2 to plus/minus 10

 

      percent.  Anything beyond 10 percent is considered

 

      a level 3 change.  Again, this is for

 

      non-release-controlling excipients.

 

                If you go down to release-controlling

 

      excipients for modified-release products, the

 

      criteria are more stringent.  Now, the change is

                                                               110

 

      measured as a percentage of the total

 

      release-controlling excipients and not the total

 

      dosage form unit so your denominator is a smaller

 

      number.  The percentage allowed is smaller for

 

      release-controlling excipients.

 

                For level 1 change, that means that the

 

      total additive effect of all release-controlling

 

      excipients should not be more than plus/minus 5

 

      percent.  Level 2 should not be plus/minus 10

 

      percent.  Changes beyond plus/minus 10 percent are

 

      considered level 3.

 

                So, this is a summary of what we have

 

      recommended in the SUPAC-IR and MR guidances.

 

      These guidances define the tests; filing document

 

      recommendations; level of changes in composition

 

      and components, release-controlling and non-release

 

      controlling excipients; site changes; batch size

 

      changes; equipment and process changes.

 

                The following changes either need a bio.

                                                               111

 

      study or an established IVIVC:  Level 3

 

      release-controlling and level 3 non-release

 

      controlling change; level 2 release-controlling

 

      change for NTR drugs; and level 3 site change and

 

      level 3 process change.  All of those changes,

 

      meaning level 2 changes, would require comparable

 

      dissolution documentation, meaning, as I said,

 

      profile comparison in several media.

 

                As I mentioned in the title slide for

 

      these guidances, the equipment addendum came out a

 

      little later and there we identified equipment by

 

      class and subclass for all major unit operations,

 

      and a change to a different class is generally

 

      considered a change in design and principle.  So,

 

      if you have equipment changes within the same

 

      design and operating principle it is considered a

 

      minor change.  If you go to a different design and

 

      principle it is a major change.  Finally, the

 

      changes guidance allows for multiple different

 

      level changes.  As we all know, these changes do

 

      not occur only one at a time; it is a composite of

 

      changes for any change.  So, if you have, say,

                                                               112

 

      several level 1 changes and one level 2 change for

 

      your new product you would be held to the most

 

      restrictive individual change of level 2, and

 

      whatever requirements go with that level of change.

 

                So, that was a quick overview of the

 

      guidances.  These documents are available on the

 

      web, and if you have any questions please look them

 

      up.  Let me switch gears here and take you through

 

      some examples of the way the specifications are

 

      set.

 

                But before that, let me share with you

 

      generally what we see in an application in terms of

 

      information available for setting specifications.

 

      The data that are available for a typical

 

      immediate-release product in an NDA are as follows:

 

      Dissolution results under a variety of agitation

 

      and media conditions.  Then typically what we see

 

      are several methods.  One method is selected by the

 

      sponsor which generally provides you with a rapid

 

      dissolution profile.  Using that method, we have

 

      data of 6-12 units and that is the limit of data we

 

      have for any given lot.  So, that is the range of

                                                               113

 

      variability that you would typically see for a

 

      particular lot.  Using that method, you have

 

      dissolution data from the bio. batch, the batch on

 

      which bioavailability has been characterized, plus

 

      few to several production lots under this

 

      condition.  Again, as I said, these batches are

 

      usually in very large quantities, hundreds of

 

      thousands to million units.  We see the data on

 

      6-12 units.

 

                Then we do have a lot of bioavailability

 

      data on this product.  Actually, bioavailability,

 

      relative bioavailability, bioequivalency trials and

 

      dissolution data of lots used in efficacy trials

 

      and stability data.  So, we look at all this

 

      information and try and come up with a meaningful

 

      specification.

 

                What do we do when we consider setting

 

      specifications?  These are the factors that are

 

      taken into consideration when setting specs. for an

 

      immediate-release product.  The in vivo behavior of

 

      a drug product, particularly how rapidly the drug

 

      is absorbed and an indicator for that is Tlag time

                                                               114

 

      or what is the Tmax of your product.  Since the

 

      issuance of the BCS guidance we look at the

 

      permeability data very closely.  In vivo

 

      permeability would be based on mass balance studies

 

      as well as absolute bioavailability studies and

 

      that, in my mind, is the gold standard by which you

 

      define whether a drug is highly permeable.  If it

 

      is quantitatively absorbed, then you say this high

 

      permeability, along with your high solubility data,

 

      puts the product into BCS class 1.  Then that

 

      carries its own benefits.  I have an example of

 

      that to show you a little later.

 

                That is what one pays attention to, in

 

      vivo behavior of the drug product from a

 

      bioavailability point of view.  We look at

 

      dissolution behavior across all conditions in vitro

 

      and then we try to come up with an adequately

 

      discriminating method, taking all this data into

 

      consideration based on any quantitative or

 

      qualitative in vitro inference.

 

                What is very helpful for evaluation of an

 

      NDA is if you have data like this where a solid

                                                               115

 

      dosage form in vivo is compared to something that

 

      is even more rapidly dissolving, meaning your solid

 

      dosage form's performance in vivo with respect to,

 

      like, a solution.  If we have this data, this tells

 

      us a lot about what is the in vivo dissolution of

 

      your solid dosage form and that can help us

 

      evaluate the in vitro considerations for setting

 

      specifications for that product.  So, this can

 

      guide how discriminating the in vitro method needs

 

      to be.

 

                As I said, we look at all the available

 

      dissolution data and pay particular attention to

 

      the lots that have in vivo data, and then discuss

 

      with our chemist colleagues about what is available

 

      in the stability domain, the data there and the

 

      specifications we are considering.  If we see a

 

      significant change or time with stability

 

      performance, that will have to be resolved by a

 

      bioequivalency study.

 

                Possible outcomes in terms of setting

 

      specifications, one is everybody is happy.

 

      Sufficient data are submitted and specs are

                                                               116

 

      finalized.  It is possible that insufficient data

 

      are submitted.  Based on the product's indication,

 

      the product needs to be approved with reset interim

 

      specs.  We agree with the sponsor what additional

 

      data needs to be generated.  We agree upon a

 

      time-line.  We evaluate the specs and we finalize

 

      the specs.  In the rare instance where there is

 

      insufficient data submitted--I have not seen this

 

      happen in my lifetime where we have withheld

 

      approval for a drug product because of insufficient

 

      dissolution data.  At the least, we will set specs

 

      on the clinical trial product.  So, if insufficient

 

      data are submitted and specs can't be finalized

 

      even including interim specs, then we have to

 

      resolve that prior to approval.

 

                Now let me take you through some specific

 

      examples, starting with simple to a little bit more

 

      complex.  This is an immediate-release drug product

 

      A.  The drug is highly soluble over the pH range of

 

      1.2-6.8, or 6.9 in this case.  Based on the

 

      bioavailability and the in vitro permeability, we

 

      established that the drug is highly permeable.  So,

                                                               117

 

      we have high solubility, high permeability criteria

 

      met.  The drug product is rapidly dissolving over

 

      the pH range of 1.2-6.8.  So, we have seen this.

 

      We are sure of these characteristics and we say

 

      okay, this is BCS class 1.

 

                We have dissolution results of the

 

      bioavailability lot and the clinical lot so all

 

      that data is utilized in setting the

 

      specifications.  There was stability data also

 

      available that was taken into consideration.  It

 

      turned out to be a straightforward case.  The

 

      sponsor's proposal was that they use a USP 1

 

      apparatus at 100 rpm in 900 ml 0.1 normal

 

      hydrochloric acid; specs of 80 percent in 30

 

      minutes.  We agreed with the sponsor.

 

                Just as a note, Ajaz and I didn't exchange

 

      notes beforehand but in this case the sponsor chose

 

      apparatus 1 to avoid coning effect.  Ajaz had an

 

      example from the Canadian database where that was

 

      the reason why you saw a big investigator

 

      difference compared to the reference, but the in

 

      vivo data turned out to be fine.

 

                Another example for an immediate-release

 

      drug product, product B, the drug is a free base

 

      with 2 pKs of 5.4 and 7.2.  It is highly soluble at

                                                               118

 

      pH 1 but it is practically insoluble at pH 7, and

 

      the solubility drops sharply between pH 4-5.  I

 

      have a graph that shows that clearly.  The drug is

 

      absorbed slowly, at Tmax ranging from 3-5 hours.

 

      The half-life is long, 45 hours.  It is not highly

 

      permeable.  The fraction absorbed is around 0.75.

 

                So, what do we do with this?  This is the

 

      dissolution behavior across the pH ranges.  As you

 

      see, below pH 5, which is the third curve from the

 

      top, dissolution starts dropping rapidly as the pH

 

      increases.  The sponsor chose the dissolution

 

      method at pH 5, and showed that the clinical and

 

      to-be-marketed formulations had similar profiles.

 

                This is what that comparison is at pH 5.

 

      We had bioequivalency data on these two

 

      formulations and that turned out to be clearly

 

      bioinequivalent in vivo for the test, meaning that

 

      to-be-marketed product showed a clear difference in

 

      Cmax.  The Cmax was 17 percent lower.  We

                                                               119

 

      interacted with the sponsor and they optimized

 

      their method to come up with an adequate

 

      discrimination condition to evaluate this

 

      formulation further.

 

                This is what they came up with, 5 percent

 

      volume Tween 80 and the same two formulations that

 

      were clearly bioinequivalent in vivo, they were

 

      able to identify their in vitro performance and

 

      show that, indeed, they were different.  This was

 

      verified further by taking the two formulations

 

      that were bioinequivalent in vivo and the method

 

      showed that they were similar in vitro.

 

                This was the availability of dissolution

 

      data across several batches.  All I want to point

 

      out to you is that, as I said, dissolution data for

 

      different batches, from 6 units, mean and range is

 

      available and if we look at the right-hand column,

 

      the lowest range is 86 percent.

 

                Taking all that data into consideration,

 

      the sponsor proposed the specification with

 

      apparatus 2 at 50 rpm and 1000 ml to Tween 80 in

 

      water; Q of 75 percent in 45 minutes.  We

                                                               120

 

      recommended no changes in the condition but a Q of

 

      80 percent in 45 minutes.  Here is an example of

 

      availability of in vivo data optimizing the

 

      specifications.

 

                The final example I have is for a drug

 

      product, a modified-release drug product with in

 

      vivo/in vivo correlation.  For this drug product a

 

      level A correlation was established.  Correlation

 

      was obtained from in vivo data from 6 different

 

      studies, and the media consisted of pH 1.5 for the

 

      first 1.5 hours and then pH 6.8 for the remainder

 

      of the 24 hours.  This is a once a day product.

 

                These are the results.  I think this was

 

      excellent work on the sponsor's part.  We worked

 

      with them and we were very happy to figure out the

 

      specs with them.  Look at the hatched region.  That

 

      is the observed range of dissolution data.  That is

 

      the extent of variability across the entire

 

      manufacturing experience for this sponsor.  So, the

 

      hatched area is the dissolution variability,

 

      dissolution range the product showed in vitro.  The

 

      specs we agreed upon are the two dotted lines above

                                                               121

 

      that hatched region.  So, those were the

 

      specifications proposed and we agreed with them.

 

                The best part is that if you look at the

 

      third level of curves, which are the topmost dotted

 

      lines, the topmost and the bottom, those are the

 

      predicted in vitro dissolution behaviors of two

 

      formulations that would be comparable in vivo.  So,

 

      the specifications were set within the limits of

 

      what products would be bioequivalent, so a good

 

      IVIVC that could lead to meaningful specifications.

 

                Now let me conclude with some personal

 

      comments on opportunities for improvement.  Before

 

      I get into my own suggestions, I want to cite this

 

      article that Ajaz already mentioned from Dr. Janet

 

      Woodcock, a clinician who has written beautifully

 

      on pharmaceutical quality.  I am just going to cite

 

      two quotations out of this article.  I mean, I can

 

      stand here and tell you a great deal about all the

 

      complexities involved in clinical trials but I

 

      think Dr. Woodcock has summarized this very well in

 

      this first bullet, which is, as she says, for the

 

      purposes of clinical use, the established drug

                                                               122

 

      quality attributes are generally adequate because

 

      they achieve much tighter control of the level of

 

      variability than could be detected in patients

 

      without extensive study.

 

                These are part of all the variabilities,

 

      specially manufacturing variability.  It can be

 

      done but it is a difficult task and it would be

 

      very extensive, and that is not the paradigm

 

      currently used.

 

                But maybe even more important, as she

 

      points out here in the very second line of the

 

      previous quotation, in contrast, for regulatory and

 

      manufacturing processes, the lack of detailed

 

      understanding of the real-world importance of

 

      quality attributes is a serious problem, leading to

 

      many disputes that might be resolved easily were

 

      relevant information available on the relationships

 

      between various quality parameters and clinical

 

      performance.  I personally couldn't agree more with

 

      that concluding comment.

 

                So, clinical performance, if I were to

 

      dissect that further--everybody talks about

                                                               123

 

      variability and this is my share of what are the

 

      different types of variability in therapy.  You

 

      start with manufacturing variability, then you have

 

      variability associated with the drug exposure and

 

      then you have variability associated with the drug

 

      response.  You have compliance issues.  You know, a

 

      lot of people can actually add more bullets to this

 

      and provide a complete picture of how complex the

 

      system is when a patient is being treated in vivo.

 

                But I have taken a shot at just making a

 

      point on exposure-related variability and

 

      manufacturing aspects associated with that.  The

 

      next table is a snapshot.  We have an internal BCS

 

      database of almost 200 NDAs.  That is in the

 

      process of being audited and we hope to publish

 

      that soon.  So, what I requested Dr. Uppoor to do

 

      is to randomly select a few drugs and prepare a

 

      table that would show variability in AUC and Cmax

 

      and the exposure parameters of different BCS

 

      products.

 

                Again, this is tentative because this is

 

      not fully audited so that is why I have starts in

                                                               124

 

      this table.  This is BCS class 1, 2, 3 and 4 across

 

      the top horizontal line.  You have the permeability

 

      associated with the AUC parameter and the Cmax

 

      parameter for these products.  As you can see,

 

      staring with class 1, we have variability in the

 

      range of 17 to about 24 percent.  Class 3 shows

 

      maximum in vivo variability.

 

                So, if I want to take this tentative class

 

      information further, the point I want to make--the

 

      numbers might be off when we have the actual

 

      publication coming out, but this is the point I

 

      want to make, that if I assume that the clinical

 

      trial formulation for this product was

 

      optimized--if it is not optimized, I think it is in

 

      the interest of the sponsor to optimize that so

 

      that even a little bit of manufacturing variability

 

      does not reflect in the in vivo performance at

 

      least from a drug exposure point of view.  But

 

      assuming that this formulation is optimized, even

 

      for BCS class 1 products you do see a decent amount

 

      of variability in vivo.  Again, this is reflecting

 

      how the drug is handled by an individual and the

                                                               125

 

      variability of handling that across individuals.

 

      This information can be utilized by a sponsor to

 

      come up with rational specs.

 

                These are some of my thoughts in terms of

 

      opportunities for improvement.  The first point is

 

      nothing earth-shattering but I still think it is a

 

      point that has to be made, to select an appropriate

 

      dissolution method based on physicochemical in

 

      vitro and in vivo characteristics of the drug and

 

      the drug product.

 

                It would be useful to have an estimate of

 

      in vitro variability for low solubility and low

 

      permeability.  Estimate of variability of lots used

 

      in pivotal efficacy trials would facilitate setting

 

      of rational specifications.  For modified-release

 

      products estimate the in vitro release

 

      variability--the example I showed where if you had

 

      a handle on the variability across your entire

 

      manufacturing process, then you can bring that into

 

      setting a meaningful specification.  As I already

 

      mentioned, right now the IVIVC current guideline is

 

      based on the limit mean estimates only and if you

                                                               126

 

      can build in the variability aspect and in vivo

 

      performance based on estimate of mean as well as

 

      variability, I think that would lead to more

 

      rational specs, maybe even wider specs compared to

 

      what we are doing now.

 

                The things that I see in the near future

 

      are new technologies like PAT.  Hopefully, it can

 

      provide in vitro and in vivo relationships based on

 

      the performance of an individual dosage form unit.

 

      I mean, this would be a non-destructive method.

 

      You would be able to assess the dissolution

 

      performance of a unit without breaking it up and

 

      then you would administer that to an individual and

 

      you would get that individual's exposure parameters

 

      so you would have correlation relationship on an

 

      individual dosage unit form in an individual

 

      patient taking it.  I think that would be a very

 

      powerful set of data to set meaningful

 

      specifications.

 

                We are getting more and more complex

 

      products like drug eluting stents and liposomes.

 

      For these complex dosage forms I think it would be

                                                               127

 

      essential to study drug elution, drug release using

 

      mechanistic models and new techniques in imaging

 

      and fluid dynamics.  Hopefully, future

 

      specifications will be based on in vitro mean and

 

      variability estimates.

 

                Moving from a science point to a process

 

      point--I didn't know our good friend Dr. Chuck

 

      Hoiber [ph.] would be here but this is in those

 

      days when Chuck and I were on the same floor and we

 

      started implementing this which is that from the

 

      process point of view there are also a lot of

 

      opportunities to optimize setting of specifications

 

      and that, from my perspective, is come and meet

 

      with us early.  A meeting would be useful if you

 

      have good quantity and quality of data.  As we have

 

      done on several occasions, we have had separate end

 

      of Phase II meetings with CMC Biopharmaceutics and

 

      colleagues on our side and the industry, going over

 

      the development plan and that has led to a quicker

 

      review and arriving at meaningful specifications at

 

      the time of NDA approval.

 

                Finally, I do personally believe that good

                                                               128

 

      homework will always bring dividends.  If you have

 

      good data, please share them with us and we will

 

      work with you to come out with rational

 

      specifications.  Thank you.

 

                     Questions by Committee Members

 

                DR. COONEY:  Thank you very much.  Some

 

      questions from the committee?  Ken?

 

                DR. MORRIS:  Two things.  I was a little

 

      surprised to see the high variability with BCS 3.

 

      In principle, you would expect BCS 3 to be a good

 

      candidate for waiver because, as long as your

 

      driving force doesn't change, you would expect that

 

      the absorption is rate limiting and falls into the

 

      same basic concept as 1.

 

                DR. MEHTA:  That is a very good

 

      observation.  We are looking at the data carefully

 

      ourselves, but I think it is maybe one product that

 

      is--

 

                DR. MORRIS:  Driving the variability?

 

                DR. MEHTA:  Yes.

 

                DR. MORRIS:  Or is it that the absorption

 

      itself is just variable?

 

                DR. MEHTA:  Again, we can think about it

 

      but it is a question if you have a class 3 high

 

      solubility, low permeability drug and if low

                                                               129

 

      permeability is not leading to the same conditions

 

      in vivo that is going to take away some of your

 

      high solubility benefit.

 

                DR. MORRIS:  Not the same conditions on

 

      which side?  Are you talking about in the gut?

 

                DR. MEHTA:  Yes.

 

                DR. HUSSAIN:  Sorry, if I may, I think one

 

      of the challenges is that this was always a

 

      question when we were deliberating the BCS

 

      guideline, high solubility.  But the in vivo

 

      dissolution actually is more sensitive for low

 

      permeability drugs and we actually have published

 

      on this with Lawrence--

 

                DR. MORRIS:  Right.

 

                DR. HUSSAIN:  So, people often say this is

 

      high solubility so dissolution is not rate limiting

 

      but in vivo dissolution behavior is quite complex.

 

      Plus, you add site-specific absorption of these

 

      compounds that adds to all the sources of

                                                               130

 

      variability.

 

                DR. MORRIS:  Right.  I guess that is my

 

      point in a sense.  Shouldn't the compounds be

 

      segregated into site-specific and passive absorbed

 

      compounds to really do a valid experiment?

 

                DR. HUSSAIN:  I fully agree with you.  We

 

      came up with the classification system and those

 

      four classes are beautiful but there is nothing

 

      that black and white.  Greater than 90 percent

 

      permeability, highly permeable, but there is a

 

      gradation of that and, you know, we have to take

 

      that into account.  You know, there are, like,

 

      windows of absorption.  So, we need to subclassify

 

      those four classes and then come up, you know, with

 

      better--

 

                DR. MORRIS:  Yes, but it would be nice if

 

      you could identify some more waiver-worthy classes.

 

                DR. HUSSAIN:  Yes.

 

                DR. MORRIS:  Just another quick comment is

 

      that I am sure it won't surprise you but, you know,

 

      with the general BA/BE guidance people, because of

 

      what is in the guidance, are actually doing pH

                                                               131

 

      solubility profiles of non-ionizable compounds.

 

                [Laughter]

 

                DR. MEHTA:  That is taking us too

 

      seriously!

 

                DR. COONEY:  Paul?

 

                DR. FACKLER:  I have a comment and a

 

      question.  The comment had to do with the slide

 

      where you suggested there might be about 20 percent

 

      variability for even BCS class 1 compounds.  I

 

      would suggest that that is vastly understated, that

 

      the variability is much higher than that because I

 

      am guessing that your data comes from

 

      bioequivalence studies where all of the subjects

 

      take exactly the same amount of water, the same

 

      amount of food.  None of them are BMI greater than

 

      a particular number.  If they are old studies they

 

      were all men.  I would just say that in the general

 

      population with the way pharmaceuticals are really

 

      taken--some people run three miles, come home and

 

      then swallow their tablets; some people roll out of

 

      bed and swallow them without water--the variability

 

      even for class 1 is significantly higher than 20

                                                               132

 

      percent.  But it is just my opinion.

 

                DR. MEHTA:  That would just add to the

 

      thought I had which is, you know, use that

 

      information to evaluate your in vitro

 

      specifications.  That will help you.

 

                DR. FACKLER:  The question I had had to do

 

      with that same chart where you looked at 17 drugs

 

      that were randomly pulled out of the pool of 200.

 

      It was interesting to see that the class 3 is N

 

      equals 7.  I am just wondering if the distribution

 

      of these 17 in any way represents the distribution

 

      of the 200 drugs.

 

                DR. MEHTA:  I don't think so.  The whole

 

      idea was to see if we can get a handle on what is

 

      the exposure variability for these products.  A few

 

      years ago I presented this database at one of the

 

      APS workshops what was surprising is that we saw a

 

      lot of NDAs falling into class 4 category.  If it

 

      is a class 4, then you would see very few drugs,

 

      low solubility, low permeability.  You know, they

 

      would fall out of drug development.  But, as I

 

      mentioned a little while ago, the way classifying

                                                               133

 

      we have created these four classes, 90 percent of

 

      data goes in class 1 over this 85 percent

 

      absorbed--you know, it is still low permeability.

 

      So, I don't think when we come out with this

 

      information, all audited, that there is going to be

 

      a majority of them falling in class 3.  I don't

 

      think so.

 

                DR. COONEY:  When you presented the table

 

      of the 17 samples, your intent is to expand that?

 

      This is just a piece of work in progress?

 

                DR. MEHTA:  Yes, very much so.

 

                DR. COONEY:  So, the idea is to really

 

      address the question that was just asked, that is,

 

      to have an analysis that is representative of that

 

      whole set?

 

                DR. MEHTA:  Yes.  I mean, right now we are

 

      going through each drug and making sure, to our

 

      level best effort, that the data available

 

      classifies that drug product in the appropriate

 

      class.  We have the information put together and

 

      now it is like careful auditing going on.

 

                DR. COONEY:  Good.  Marvin?

 

                DR. MEYER:  I did come up with a couple of

 

      questions.  It always bothered me that the BCS

 

      system had this quadrant drawn and then the lines

                                                               134

 

      kind of floated depending on how you wanted to

 

      define high and low--

 

                DR. MEHTA:  No, it is rigid right now.

 

                DR. MEYER:  I know it is rigid but the

 

      rigidness was arbitrary.

 

                [Laughter]

 

                It is arbitrarily rigid.

 

                DR. HUSSAIN:  I will defend it tomorrow;

 

      don't worry!

 

                DR. MEYER:  Okay.

 

                DR. MEHTA:  We started out with a

 

      conservative position and now with the availability

 

      of more data we want to expand that rationally with

 

      proper evidence.

 

                DR. MEYER:  It also bothered me that this

 

      permeability goes all the way from a very rigorous

 

      intubation of humans to a K2 cell to looking at

 

      Tmax.  So, how it is defined or determined can be

 

      another source of variability in where it falls in

                                                               135

 

      this rigorously arbitrary quadrant.  So, I think

 

      that may be a reason in part why the class 3 seemed

 

      to be more variable than 2.  One drug in that would

 

      have expanded the range.

 

                DR. MEHTA:  That is just the way those

 

      drugs got pulled out.  That is why I have that

 

      range.  That may not be reflective of what it is.

 

      I don't want to take up too much time, but we look

 

      at permeability assessment now very carefully and,

 

      in my mind, hopefully, if we have data on the NDA

 

      side, which is mass balance data and

 

      bioavailability data, that is the maximum way in

 

      terms of assessing, you know, whether the drug is

 

      90 percent absorbed or not.  Sometimes we have an

 

      issue with that.  Then we utilize the in vitro

 

      methods for that decision.

 

                DR. MEYER:  One last question.  Do you

 

      feel that the f2 test has been rigorously

 

      evaluated?

 

                DR. MEHTA:  A good question, Marv.

 

                [Laughter]

 

                There are people in the audience that--

 

                DR. MEYER:  Do you feel--do you feel it

 

      has been rigorously evaluated so it will detect

 

      differences when they should be detected and will

                                                               136

 

      allow passage when it should be allowed?

 

                DR. MEHTA:  Well, I mean we do state in

 

      our guidances under what conditions this approach

 

      should be employed.  You know, if your variability

 

      is very high in dissolution on each formulation

 

      this is not the right way of comparing those

 

      profiles so then you need to get into more complex

 

      assessment, and all that.  If it is done properly,

 

      yes, I do myself.

 

                DR. COONEY:  Pat?

 

                DR. DELUCA:  In the BA/BE guidance summary

 

      for modified-release products you are saying that

 

      they should profile using at least three other

 

      dissolution media and water.  Why do you need three

 

      others if you have a correlation?

 

                DR. MEHTA:  No, it doesn't say that there

 

      is a correlation.  This is just a question--well,

 

      usually correlation is release formulation

 

      specific.

 

                DR. HUSSAIN:  It is just for that product.

 

      So.

 

                DR. MEHTA:  It is right now.

 

                DR. COONEY:  Nozer?

 

                DR. SINGPURWALLA:  When you don't

 

      understand something you start asking technical

                                                               137

 

      questions.

 

                [Laughter]

 

                You showed a picture of linear correlation

 

      long ago, one of your early slides--

 

                DR. MEHTA:  Yes, level A correlation.

 

                DR. SINGPURWALLA:  Level A correlation.  I

 

      have two comments.  The first is that you are

 

      looking for relationships between the percent of

 

      drug dissolved and the percent of drug absorbed so

 

      correlation only measures linear relationships.

 

      You may have dependence which may be not linear but

 

      still of value to you, but correlation does not

 

      measure that.  So, I just want to say that as a

 

      comment.

 

                The second more serious comment is that

 

      that particular correlation misses the time index. 

                                                               138

 

      What you really need is a third axis also showing

 

      the time at which all these happen.  For you to do

 

      that, you want to look at these two as what we

 

      would call stochastic processes or time series, and

 

      you want to cross-correlate the two time series.

 

      So, if you want to improvise on that particular

 

      theme, you may want to look not at correlation but

 

      what I would consider cross-correlation where you

 

      also introduce the time axis.  That is the only

 

      comment I want to make.

 

                DR. MEHTA:  Thank you.  That is helpful.

 

                DR. SINGPURWALLA:  Do you want to

 

      challenge me now?

 

                DR. MEHTA:  No, I didn't say that.

 

                DR. COONEY:  Are there any other questions

 

      at this point?

 

                [No response]

 

                Thank you.  The next presentation will be

 

      by Dr. Shah establishing dissolution

 

      specifications.

 

                Establishing Dissolution Specifications:

 

                            Current Practice

 

                DR. SHAH:  Good morning.  Mehul gave a

 

      nice overview on the BCS guidance and other

 

      guidances which are used in setting dissolution

                                                               139

 

      specification from a biopharmaceutics perspective.

 

      My job today is to cover the CMC aspects of setting

 

      the dissolution specifications.  In this

 

      presentation I am going to start with an overview

 

      of the current practice, and in that overview I am

 

      going to cover the CMC assessment and bring in some

 

      of the ICH Q6A principles, how we evaluate the ICH

 

      Q6A principles in our CMC assessment, and then I

 

      would like to talk about a case study example for

 

      extended-release oral suspension and in that

 

      example I am going to cover the drug development

 

      strategy by the applicant, the dissolution results

 

      obtained based on that development strategy, then

 

      what we identified as critical issues, followed by

 

      our recommendations and based on those

 

      recommendations, what were the improvements

 

      implemented by the applicant and what was the

 

      outcome out of those implementations.  I would like

 

      to end my talk with some concluding remarks based

                                                               140

 

      on this example as well as general remarks in that

 

      aspect.

 

                As Mehul suggested in his presentation, I

 

      want to reemphasize that establishing dissolution

 

      specification is a shared responsibility between

 

      the Office of New Drug Chemistry and the Office of

 

      Clinical Pharmacology and Biopharmaceutics.

 

                In the next three slides I have presented

 

      the considerations that should be given during that

 

      development, as well as the focus of CMC assessment

 

      during the NDA review, and what forms the basis of

 

      setting the dissolution specifications from CMC

 

      perspective.

 

                As I have pointed out here, it is a known

 

      fact that physicochemical properties of the

 

      formulation components, such as drug substance and

 

      other excipients, such as the solubility, pKa,

 

      particle size distribution, polymorphic forms and

 

      there may be some others, have a significant effect

 

      on the dissolution.  The physicochemical properties

 

      impact the processibility of the formulation

 

      components, as well they may affect also the

                                                               141

 

      safety, efficacy and stability of the drug product.

 

      In addition to that, the manufacturing processes,

 

      especially those having the potential to influence

 

      the release profile of the drug substance also

 

      should be studied during the development.  And, the

 

      control strategy of the critical process parameters

 

      and in-process testing also should be developed

 

      during the development, and those are the focuses

 

      of the CMC assessment.

 

                During the drug development one should

 

      expect that there should be a relationship of

 

      in-process testing to the critical quality

 

      attributes, such as dissolution of the drug

 

      product.  Some of the in-process testing that may

 

      be carried out might be particle size distribution;

 

      release rate; and the compression force, tablet

 

      hardness and friability in the case of solid oral

 

      dosage form.

 

                In addition, during the CMC assessment we

 

      focus also on the development and validation

 

      aspects of the proposed in vitro dissolution

 

      method.  Cindy already covered some of these

                                                               142

 

      aspects in terms of how the methodologies are being

 

      developed and what are the validation criteria that

 

      need to be covered, especially pertaining to

 

      specificity, linearity, accuracy, precision,

 

      ruggedness, etc.  In addition, we also focus on the

 

      release time point intervals and what should be the

 

      adequate tim point intervals.

 

                Once we have this information we need to

 

      see or need to provide during development, as well

 

      as the NDA submission data, what is the

 

      relationship between the in vitro dissolution data

 

      from development, clinical, bio. and primary

 

      stability batches, and also identify a discerning

 

      trend on storage.  We also evaluate the proposed

 

      shelf-life of the drug product on the basis of the

 

      stability data analysis of dissolution, as well as

 

      other drug product attributes.

 

                In the end, it is in coordination with

 

      Office of Clinical Pharmacology and Pharmaceutics

 

      that appropriate dissolution specifications are

 

      recommended and these specifications are reflective

 

      of the dissolution data from various batches

                                                               143

 

      including clinical, bio., stability and other

 

      batches.

 

                In terms of the ICH Q6A document, ICH Q6A

 

      discusses the potential relevance of particle size,

 

      polymorphic content and polymorphic changes, and

 

      how it affects the dissolution.

 

                Here I have these three decision trees

 

      just for reference.  I just wanted to point out

 

      that CMC assessment very well integrates these

 

      principles in our assessment for the quality

 

      assessment of the drug product.  This is about the

 

      particle size distribution and the decision tree

 

      guides you on how to set acceptance criteria.

 

                This is in terms of polymorphic content.

 

      That also guides you on how to set acceptance

 

      criteria.  The next one is how to set the

 

      polymorphic change acceptance criteria in the drug

 

      product.

 

                Now I would like to focus on the case

 

      study example for extended-release oral suspension

 

      for the remainder of my talk.

 

                Let me give you just some background. 

                                                               144

 

      This was submitted as a 505(b)(2) application.  As

 

      a result, there was no clinical trial required

 

      because the safety and efficacy of the proposed

 

      active ingredients for the proposed indication was

 

      established through immediate-release products

 

      available under the tentative OTC monograph for the

 

      same indication.  The proposed dose was a single

 

      dose given every 12 hours to patients 6 years of

 

      age or older.  That was equivalent to the nominal

 

      OTC monograph which was given every 6 hours twice.

 

                In terms of the formula, the drug product

 

      contained two different active ingredients, and I

 

      will call them drug substance 1 and drug substance

 

      2.  For proprietary reasons, most of the data I am

 

      going to discuss here are well concealed and they

 

      are masked but the data are real.  Drug substance 1

 

      is anchored to a drug carrier support and coated

 

      separately with semipermeable polymer to prevent

 

      dose dumping and to impart the extended-release

 

      profile.  Drug substance 2 binds the drug carrier

 

      support in situ during the manufacturing process,

 

      but it is not coated.  Both active ingredients,

                                                               145

 

      along with other excipients, are suspended in

 

      aqueous solution.

 

                The concerns we had here arise from the

 

      safety implications due to the potential dose

 

      dumping, and efficacy implications due to

 

      insufficient rate and the extent of release of the

 

      actives.  These concerns were brought to the

 

      applicant's attention during the end of Phase II

 

      meeting as well as pre-NDA meetings, and they were

 

      very mindful of those two concerns.

 

                This was the strategy adopted by the

 

      applicant in the beginning.  They wanted to

 

      demonstrate bioavailability of the drug product

 

      formulas, and that was coated with 6 percent

 

      coating of drug substance 1, to a reference drug

 

      which as an immediate-release solution, and it was

 

      containing the same two active ingredients.  They

 

      had no other choice but to start with the

 

      immediate-release solution because there was no

 

      existing extended-release product containing these

 

      two ingredients.

 

                Their plan was to formulate three

                                                               146

 

      experimental drug formulations, each differing only

 

      by the coating level of semipermeable polymer on

 

      drug substance 1.  They were low coating, for

 

      example, 2 percent; medium coating, example, 5.5

 

      percent; and high with 9 percent coating on drug

 

      substance 1.  They labeled them as fast-release

 

      solution, intermediate-release formulation and

 

      slow-release formulation.  The approach was to

 

      establish IVIVC for each active among these three

 

      experimental formulations, and establish

 

      dissolution specifications for both actives based

 

      on generated dissolution profiles from the slow-

 

      and fast-release drug product formulations.

 

                In the NDA the data submitted include five

 

      formulations of the drug product containing drug

 

      substance 1 coated with varying levels of

 

      semipermeable polymer, 2 percent, 5.5 percent 9

 

      percent, as well as 6 percent and 10 percent.  They

 

      performed the following PK studies, multi dose

 

      bioavailability studies with immediate-release

 

      solution and single dose food effect study

 

      containing 6 percent polymer coating, and single

                                                               147

 

      dose IVIVC study containing three formulations, 2

 

      percent, 5.5 percent and 9 percent polymer coating.

 

      In support, there were PK results from four batches

 

      and stability results from four PK and five

 

      stability batches.

 

                Based on these PK studies, these were the

 

      applicant's claims, that level A IVIVC was

 

      established for both actives of the ER suspension.

 

      The mean individual level A IVIVC models for drug

 

      substance 2 met the FDA validation criteria and, in

 

      their opinion, it can be used for setting

 

      dissolution specifications and biowaivers.

 

                The mean and individual level A IVIVC

 

      models for drug substance 1, which is coated,

 

      failed the FDA validation criteria in that the

 

      predicted values had a larger error than

 

      recommended.  However, if the dissolution criteria

 

      remain within dissolution profiles tested in IVIVC,

 

      they proposed that the drug substance 1 results can

 

      serve as a mapping study for the formulations.

 

                Now let's see what was the agency's

 

      finding in terms of the PK results.  On the

                                                               148

 

      bioavailability and food effect studies, which was

 

      the 6 percent coating of drug substance 1, the

 

      agency found that systemic exposures of both

 

      actives were favorable between the extended-release

 

      suspension and multi dose of reference

 

      immediate-release solution, and there was no food

 

      effect on both actives.

 

                However, in terms of the IVIVC study,

 

      where the drug substance was coated with the 2

 

      percent formulation, 5.5 percent formulation and 9

 

      percent formulation, with respect to drug substance

 

      1, the agency found that it failed to establish the

 

      in vivo/in vitro correlation, and observed more

 

      than 20 percent of difference in Cmax for

 

      formulation of fast and slow dissolution profiles.

 

                With respect to drug substance 2 that was

 

      not coated, level A IVIVC was established, however

 

      it failed to validate the IVIVC.  The formulations

 

      used in the IVIVC study were found to be

 

      bioinequivalent, that is to say the Cmax of the

 

      formulations used in the IVIVC study were different

 

      by more than 20 percent.  The proposed dissolution

                                                               149

 

      specification and the approach to set a dissolution

 

      specification based on IVIVC by mapping was found

 

      unacceptable.

 

                Now let me share the stability results

 

      analysis.  This is what we review in our CMC

 

      assessment.  What we found was contradictory

 

      release profiles observed between drug product

 

      formulations containing 6 percent and 9 percent

 

      coated drug substance.  Drug substance 2 showed

 

      more decrease in dissolution than drug substance 1,

 

      and we observed substantial decrease in dissolution

 

      at 1-hour, 3-hour and 6-hour time points for both

 

      actives from the corresponding initial values among

 

      all batches, including bio. and primary stability

 

      batches, at all storage conditions.  The decrease

 

      in dissolution was most notable at 3-hour and

 

      6-hour time points.  The decrease in dissolution is

 

      minimum at the 12-hour time point and the decrease

 

      in dissolution for both actives levels off by 9

 

      months on storage.

 

                This is displayed on this slide.  This is

 

      the dissolution results of drug substance 1.  For

                                                               150

 

      clarity purpose, I have labeled the coating for the

 

      dissolution curves.  The yellow bar shows the 6

 

      percent coating that was used in the

 

      bioavailability study.  The purple is the 2

 

      percent.  The middle one is blue, which is 5.5

 

      percent coating of drug substance 1.  The red one

 

      is the 9 percent coating of drug substance 1.

 

                Now, what I explained in the previous

 

      slide is what you can see is a decrease in

 

      dissolution profiled for all the solutions.  You

 

      would expect the 9 percent would be showing a slow

 

      dissolution compared to the 6 percent but it is

 

      quite the other way.

 

                If you look at drug substance 2, the

 

      decrease is more compared to drug substance 1,

 

      which is shown basically from the least point and

 

      at the 18 months time point.  That is more than

 

      about 20 percent decrease in dissolution over time.

 

                So, based on this analysis these were the

 

      critical issues discussed with the applicant, and

 

      they concerned the raw material controls,

 

      manufacturing processing and in-process controls

                                                               151

 

      and controls related to particle size distribution

 

      and dissolution method.

 

                I just want to point out over here that

 

      these discrepancies in the results showed that the

 

      coating process was not in control and we discussed

 

      that issue with the applicant.  They decided to

 

      reformulate the drug product and decided to abandon

 

      the idea of the IVIVC approach to set dissolution

 

      acceptance criteria; conduct PK studies on

 

      commercial scale bio. batch containing drug

 

      substance 1 at the specified target coating level,

 

      rather than a range, and compare it to the

 

      reference IR solution; manufacture additional 3

 

      pilot scale primary stability batches of the drug

 

      product containing drug substance 1 at the same

 

      specified target coating level; and propose

 

      dissolution acceptance criteria based on in vitro

 

      dissolution profiles obtained for both actives from

 

      the bio. batch.

 

                These were the process improvements

 

      implemented.  They coated the drug substance with a

 

      specified target coating level of semipermeable

                                                               152

 

      polymer; revised the coating and subsequent

 

      manufacturing processes; instituted appropriate

 

      process controls to stabilize binding of both

 

      actives to the drug carrier support in the

 

      suspension; and manufactured one commercial scale

 

      bio. batch and three pilot scale stability batches.

 

                They instituted appropriate particle size

 

      measurement method, for example laser diffraction,

 

      for drug carrier support and coated drug-bound

 

      carrier particles.  They revised particle size

 

      distribution acceptance criteria for the drug

 

      carrier support, coated drug substance bound

 

      carrier support particles and suspension

 

      stabilizing excipients.

 

                Based on these results, they conducted

 

      three PK studies utilizing the drug product

 

      formulation with coating of drug substance 1.  They

 

      conducted BA/BE assessment; PK at steady state; and

 

      food effect studies.  The results showed that the

 

      PK profiles of drug substance 1 and drug substance

 

      2 from test extended-release suspension were found

 

      comparable to the reference IR solution following

                                                               153

 

      single and multiple dose administration, and food

 

      had no effect on bioavailability of both actives.

 

                Now let me share with you the stability

 

      results analysis.  After the implementation of the

 

      improvements in manufacturing process for coating

 

      and instituting adequate process controls in terms

 

      of particle size, we observed stable and consistent

 

      release profiles at 1-hour, 3-hour, 6-hour and

 

      12-hour time points for both drug substance 1 and

 

      drug substance 2 on storage within each of the bio.

 

      and three primary stability batches.  There was no

 

      discernible trend in release profiles of drug

 

      substance 1 and drug substance 2 and on bio. and

 

      primary stability batches at all storage

 

      conditions.  And, there were comparable release

 

      profiles for both drug substance 1 and drug

 

      substance 2 among bio. and three primary stability

 

      batches.

 

                This is displayed in this graph for drug

 

      substance 1.  You can see, as opposed to the

 

      dissolution rates that we saw before and after

 

      implementation of manufacturing processes.  This is

                                                               154

 

      with respect to drug substance 1, which was coated.

 

      This is the bio. batch and these are the three

 

      primary stability batches.  Most of the

 

      dissolution, as you can see, ranges between 5-7

 

      percent.

 

                This is with respect to drug substance 1

 

      dissolution profile.  This is the bio. batch and

 

      you can see these are the three primary stability

 

      batches and you do not see any discernible trend

 

      and most of the dissolution ranges between 5

 

      percent if you compare it to drug substance 2 prior

 

      to the implement.

 

                Then I would like to conclude my

 

      presentation with the following remarks.  We were

 

      able to identify probable causes of discrepant and

 

      inconsistent dissolution results for drug substance

 

      1 and drug substance 2, and recommend corrective

 

      measures to address the issues.  The outcome was

 

      consistent manufacturing process; acceptable BA/BE

 

      results; stable and consistent release profiles

 

      without any discernible trend on storage for both

 

      drug substance and drug substance 2.  Dissolution

                                                               155

 

      criteria which were set were better reflective of

 

      the data.  There was a substantial improvement in

 

      the quality of the drug product and there was a

 

      significant improvement in assurance of the safety

 

      and efficacy concerns.

 

                However, the case study example

 

      highlighted two significant points.  There was a

 

      lack of or poor understanding of the raw material

 

      properties and manufacturing processes that were

 

      critical to be controlled for consistent quality

 

      and thereby desired performance, for example,

 

      extended-release dissolution of the drug product.

 

      It also identified inadequate efforts invested by

 

      the applicant during the drug development to

 

      understand the causal links of dissolution

 

      failures.

 

                The case study example stresses a dire

 

      need for improvement to the existing drug

 

      development efforts to understand the relationship

 

      between the raw material properties of formulation

 

      components and critical quality attributes of the

 

      drug product; the effect of raw material properties

                                                               156

 

      of formulation components on their processibility

 

      for selected manufacturing processes, and the

 

      effect of manufacturing processes and associated

 

      critical process parameters on the critical quality

 

      attributes of the drug product.

 

                I would like to end my talk with the last

 

      remark that there is no substitute to a systematic

 

      and scientific approach to drug development for a

 

      safe, efficacious and quality drug product.  Thank

 

      you.

 

                     Questions by Committee Members

 

                DR. COONEY:  Thank you.  There is an

 

      opportunity for questions.  Nozer?

 

                DR. SINGPURWALLA:  Just a point of

 

      information, you repeatedly used distribution,

 

      particle size distribution.  What particle size

 

      distributions do you use in your activities?

 

                DR. SHAH:  I am not following the

 

      question.

 

                DR. SINGPURWALLA:  Particle sizes are

 

      random.

 

                DR. SHAH:  Correct.

 

                DR. SINGPURWALLA:  They are not the same.

 

      So, they have a probability of distribution.

 

                DR. SHAH:  Yes.

                                                               157

 

                DR. SINGPURWALLA:  Now, there is a lot of

 

      literature, perhaps not in your business, on what

 

      should be the distribution of particle sizes.  This

 

      morning we heard the viable distribution attacked

 

      by my colleague here, but the log normal

 

      distribution is often used as a distribution of

 

      particle sizes.  My question is what distributions

 

      are used in the pharmaceutical industry for

 

      particle sizes, or is this a completely different

 

      scenario?

 

                DR. SHAH:  I am not sure how to answer

 

      that question, but I will tell you what we practice

 

      in CMC review.  We ask for the applicant to

 

      identify the particle size range in D10, D15 and

 

      D90.  That means 90 percent of the particles--

 

                DR. SINGPURWALLA:  Right.

 

                DR. SHAH:  And we ask for the span,

 

      basically the ratio of D10 to D90 divided by D15

 

      and that gives you where the distribution lies. 

                                                               158

 

      Basically, that kind of gives control of

 

      consistency of the particle size distribution.

 

                DR. SINGPURWALLA:  Actually, you answered

 

      my question.  What seems to be not there in your

 

      industry is you are just looking at the percentiles

 

      and if the distributions are skewed one way or the

 

      other it makes a big difference what they are when

 

      you simply work with the percentiles.  So, I am

 

      just encouraging you to look into that.

 

                DR. SHAH:  I agree.  Thank you.

 

                DR. COONEY:  Ken?

 

                DR. MORRIS:  I think one of the things

 

      that occurs is that people don't control the

 

      distributions.  They tend to be log normal sort of

 

      in a general sense but people don't intentionally

 

      control this.  They usually control to a mean,

 

      which is a real big problem--

 

                DR. SINGPURWALLA:  If you control the mean

 

      you start to control the distribution.

 

                DR. MORRIS:  Yes, you try to control the

 

      mean but there is no real--and I am not sure what

 

      historically the reason is for that but that is

                                                               159

 

      sort of the case.

 

                DR. SINGPURWALLA:  You need to know what

 

      it is.

 

                DR. MORRIS:  But you need to know what it

 

      is.

 

                DR. SINGPURWALLA:  You can't control it.

 

                DR. MORRIS:  That is right.  My question

 

      is do you think that there problem was control

 

      simply of film thickness or was it perhaps

 

      incorporation of one of the compounds into the film

 

      unintentionally during the coating process?

 

                DR. SHAH:  No, that was definitely the

 

      coating process, and this was like black art in

 

      that they were mixing and matching and they never

 

      had a handle on the coating process itself.

 

                DR. HUSSAIN:  One point that I think I

 

      wanted to illustrate with this presentation was

 

      that really to control, to achieve a state of

 

      control, and so forth, you have to get down to

 

      upstream activities, starting with raw materials,

 

      and so forth.

 

                The point I also wanted to sort of

                                                               160

 

      emphasize was that just focusing on a test, even

 

      when you have a correlation, which is just a

 

      correlation and may not be causal, I think is that

 

      gap that we are also trying to fill with focusing

 

      on the CMC part of the manufacturing controls.

 

      Without that the system really--the method is

 

      weakened.  So, the quality by design aspect is to

 

      emphasize that part of it.  So.

 

                DR. COONEY:  I think another dimension

 

      with this particular case is that there is a

 

      significant amount of complexity because you are

 

      dealing with multiple products, complexity both in

 

      the process as well as in the product itself.  This

 

      is I think a particularly good example where

 

      quality by design can have a greater impact with

 

      these more complex processes and products, and the

 

      processes and the products need to be thought

 

      through together, which is your point.  It is very

 

      clear.

 

                I think we are actually going to begin

 

      lunch ten minutes early.  However, beginning lunch

 

      ten minutes early does not mean that you get an

                                                               161

 

      extra ten minutes for lunch.  We will reconvene at

 

      12:50--guess what, you can get an extra ten minutes

 

      for lunch.  We will reconvene at one o'clock.

 

                [Whereupon, at 11:50 a.m., the proceedings

 

      were recessed for lunch, to resume at 1:00 p.m.]

                                                               162

 

                A F T E R N O O N  P R O C E E D I N G S

 

                DR. COONEY:   If I could have people's

 

      attention, welcome back from lunch.  I hope that

 

      everyone appreciated the extra 9.5 minutes that you

 

      had for lunch.  It is one o'clock.  It is the

 

      opening period for open public hearing.  We have

 

      one presentation for this afternoon by Will Brown

 

      from USP, and he will speak with us on USP and

 

      dissolution testing.  Thank you.  Welcome

 

                          Open Public Hearing

 

                DR. BROWN:  Thank you so much, and I would

 

      like to thank the various FDA staff members for

 

      giving a staff member at USP the opportunity to

 

      speak before this committee.  I am a member of the

 

      staff of the Department of Standards Development at

 

      USP, and I serve as one of the liaisons to the

 

      Biopharmaceutics Expert Committee.

 

                This is breaking news.  USP reorganizes

 

      itself once every five years, and part of that

 

      reorganization is the election of the chair of the

 

      Council of Experts.  We have a reelected chair,

 

      Thomas Foster, for the Biopharmaceutics Expert

                                                               163

 

      Committee.  You can see on this slide the

 

      membership, and you will see names you recognize

 

      hopefully.

 

                USP and dissolution--well, we are terming

 

      dissolution one of the performance tests.

 

      Performance tests currently mean dissolution or

 

      disintegration test, and by test I mean part of the

 

      specification.  The ICH definition, and it is very

 

      easy to use terms loosely, says that a

 

      specification is a list of tests, associated

 

      procedures and acceptance criteria.  So, that is

 

      kind of the idea of the USP dissolution.  It is

 

      part of the specification.  You will find the

 

      public specification in the USP monograph.

 

                The general dissolution test is found in

 

      the general chapter, 7-11 on dissolution, and that

 

      gives a general description of the techniques that

 

      are available, with the understanding that those

 

      techniques can be modified.  We saw this morning

 

      what the modifications might represent.  They might

 

      represent the appropriate medium or agitation or

 

      apparatus as determined by the applicant and the

                                                               164

 

      FDA.

 

                Now, the study design that is embedded in

 

      the dissolution test and the analysis is in three

 

      stages.  We have a fixed number of samples tested

 

      at each stage and there are acceptance criteria

 

      again that are determined by the applicant and the

 

      agency, and then communicated to USP by what I am

 

      terming the sponsor, who is the same party as the

 

      applicant.

 

                The general approach is to test by

 

      attribute.  In other words, a product is either

 

      good or bad.  It either conforms or it doesn't and

 

      that is a fairly decent approximation and

 

      convenient for application by an independent

 

      analyst but it doesn't necessarily address

 

      underlying distributions of performance.

 

                In the USP test by attributes there is a

 

      control on the spread of the data.  By example, at

 

      the S3 level where you tested 24 units there is a

 

      limit that says that no individual unit value can

 

      be below Q-25 percent.  So, there is an

 

      acknowledgement that there may be an underlying

                                                               165

 

      distribution at least on stability.

 

                For the Biopharmaceutics Expert Committee,

 

      in this cycle the expert committee is working on

 

      revising general chapters to include performance

 

      tests by dosage form, by route of administration.

 

      The current approach to applied dissolution is

 

      typically two oral products and some transdermals.

 

      The routes of administration that USP has

 

      identified were discussed in a stimuli article in

 

      Pharmacopeia Forum, in September, 2003 and

 

      basically identified five basic routes of

 

      administration, topical dermal, gastrointestinal,

 

      mucosal, by injection and by inhalation.  It is

 

      just a way to cut the universe.

 

                The intention is to work with the FDA and

 

      industry as appropriate but to facilitate this work

 

      the Biopharmaceutics Expert Committee has asked for

 

      the formation of advisory panels, which have been

 

      formed.  They were formed in the last cycle and

 

      they are currently meeting.

 

                My general feeling is that meetings may be

 

      productive but oftentimes they are not.  I have two

                                                               166

 

      examples of meetings that are productive.  In 1993,

 

      I am told that the predecessor to this committee

 

      met and out of that ultimately, in '97, came the

 

      immediate-release and extended-release guidances

 

      that were talked about this morning.  Another set

 

      of meetings that happened in that same time frame

 

      are the meetings of the Pharmacopeia discussion

 

      group.  The Pharmacopeia discussion group includes

 

      the Japanese pharmacopeia, the  European

 

      pharmacopeia, the USP and the World Health

 

      Organization.  In the process of harmonization,

 

      there actually has been a common statement with

 

      respect to system suitability.  It doesn't talk

 

      about calibrators, however there is a provision to

 

      have national text and in the national text portion

 

      of system suitability the USP continues to describe

 

      calibrators as part of the system suitability

 

      determination.  The general chapters are currently

 

      at stage six and that information can be found in

 

      the current PF and the corresponding Japanese and

 

      European documents.

 

                I was told that I only had ten minutes so

                                                               167

 

      this presentation is briefer than I usually intend,

 

      but I would like to draw the committee's attention

 

      to possibly a useful document.  This document

 

      article by Walter Hauck and a group at USP talks

 

      about oral dosage form performance tests, new

 

      dissolution approaches.  It is in the recent

 

      Pharmaceutical Research, I think February 22.2.  It

 

      talks about an approach that has explicit

 

      hypothesis testing.  Parametric tolerance interval

 

      is involved.  It gives an improved way, or at least

 

      the authors believe that it is an improved way to

 

      set dissolution acceptance criteria, and allows

 

      more flexibility in the design of a protocol.  So,

 

      I will just point you at that resource.  It may

 

      have some value.

 

                It allows the industry representatives

 

      more control on study design; allows the

 

      opportunity for tiered testing.  It doesn't

 

      specifically talk about tiered testing but allows

 

      that there may be an opportunity for some kind of

 

      successive testing on failing to meet the criteria

 

      at the first level.  It allows some flexibility in

                                                               168

 

      the number of units that are tested within each

 

      tier, and it allows the possibility that the test

 

      protocol, the test design could be changed from

 

      manufacturer to manufacturer.

 

                The idea is to set a probability of

 

      passing units from a batch where the clinical

 

      properties are known.  So, you characterize the

 

      batch for in vitro dissolution; determine, in some

 

      kind of a discussion with the agency--again, I am

 

      speaking from industry perspective even though I

 

      don't represent any industry perspective--sets the

 

      fraction of the units in this idealized reference

 

      population or this actual reference population that

 

      must conform to the standard.

 

                This approach, and I won't be able to

 

      describe this more fully, the authors believe will

 

      allow the consumer and producer risks to be clearly

 

      assessed, managed and communicated.  Ideally, if we

 

      continue with the model of dissolution for

 

      performance assessment, this could be communicated

 

      publicly in the compendium.  The basic underlying

 

      approach conforms to the approach for uniformity of

                                                               169

 

      metered dose inhalers that I believe this committee

 

      will be talking about tomorrow.

 

                Finally on to calibrators, the system

 

      suitability determination is written into the

 

      general chapter and, as I interpret it, is part of

 

      the performance of any dissolution test.  So, if a

 

      dissolution test is performed for compendial

 

      purposes, currently USP requires that the apparatus

 

      is demonstrated to be suitable, and the

 

      demonstration of suitability includes successful

 

      performance of the calibrators.

 

                In actual point of fact, the use of the

 

      calibrators has a GMP function.  Test apparatuses

 

      need to be demonstrated to be suitable twice a

 

      year.  So, that is the actual application of what I

 

      believe to be more comprehensive suitability

 

      determination.  I don't currently work in the lab

 

      but when I was in the lab if there were critical

 

      dissolution experiments to be performed, they were

 

      performed on an apparatus that was calibrated

 

      before and after so that the integrity of the data

 

      was not suspect on the grounds of an unsuitable

                                                               170

 

      apparatus.  The idea of calibration is not to focus

 

      on the performance of the apparatus but to rule out

 

      unsuccessful or unacceptable apparatus, so rule out

 

      apparatus on the extremes.

 

                The extremes--there is a range of

 

      acceptable results that is determine from a

 

      collaborative study, and we try to cast the net as

 

      widely as possible so that we can capture the

 

      sources of variability in properly operating labs.

 

      Inter-laboratory variability is a major component

 

      of the ranges.  I would submit that any one

 

      dissolution apparatus or assembly, because the USP

 

      looks at the apparatus as a single vessel, single

 

      spindle combination but, in fact, we have

 

      assemblies, groups of apparatus.  So, that is part

 

      of the wideness of the range.  We can talk about

 

      that if you wish.

 

                Calibrators, what we do with calibrators,

 

      USP is aware of problems.  Salicylic acid has

 

      elegance problems.  And, we go into unit packaging

 

      in the latest batch.  Prednisone tablets, the

 

      prednisone tablets that we distribute are a

                                                               171

 

      scale-up from the University of Maryland batches

 

      that were intended to reproduce the NCDA2 10 mg

 

      prednisone tablet and we have a new batch in

 

      production.  Theophylline beads are calibrated for

 

      apparatus 3, were deleted partially in response to

 

      requests or concerns by users.   And, thank you

 

      very much.

 

                     Questions by Committee Members

 

                DR. COONEY:  Thank you.  I would like to

 

      now open up your presentation to questions from the

 

      committee.

 

                DR. MORRIS:  Just one point.  I guess if I

 

      sort of put aside for the moment the specific

 

      hesitation I have about calibrator tablets, if I

 

      just look at the criteria by which you would

 

      reproducibly generate a standard for calibration it

 

      seems to me that the things that are missing are

 

      not that unattainable.  I mean, I think that you

 

      have to have a particle size solid fraction as

 

      opposed to weight and, you know, the normal

 

      controls you have on tableting.  I don't think

 

      there is solid fraction control in the calibrator

                                                               172

 

      tablets, if I am correct.  Art, do you know?

 

                DR. KIBBE:  No, and I want to go back and

 

      have a solid cylinder without any particle size

 

      where you only have dissolution for the surface.

 

      The point that I was going to make sure I was right

 

      about is that it is my understanding that if you

 

      claim a USP product, that product must meet USP

 

      testing if USP testing is done by anybody.  But it

 

      doesn't require you to do USP testing on your

 

      product if you have a better way of controlling the

 

      quality of that product.  Isn't that right?

 

                DR. MORRIS:  That is correct.  That is

 

      clearly stated I think in the Journal of Medicine.

 

                DR. KIBBE:  Right.  So, if I was a company

 

      who was really heavy into PAT and had a really good

 

      control on my product, once I established that my

 

      product met USP guidelines for that product, I

 

      would never have to do that test again unless I am

 

      challenged.

 

                DR. BROWN:  That is correct.

 

                DR. KIBBE:  Right.

 

                DR. MORRIS:  Can I just ask is solid

                                                               173

 

      fraction controlled in the calibrator tablets?

 

                DR. BROWN:  I can't speak to the

 

      manufacturing of calibrators but I know that I have

 

      seen formulas where the solid fraction beat

 

      particle size.  My concern about a disintegrating

 

      dosage form, my personal concern is the rate of

 

      disintegration and how fast the active is exposed

 

      to the medium.  That is critical and I take your

 

      advice on the solid cylinder.  I think that we see

 

      very nice dissolution from salicylic acid tablets

 

      which, again, are being essentially remanufactured

 

      and repackaged, and I think will have the potential

 

      to show some value that we don't currently extract

 

      from them.

 

                DR. COONEY:  Tom?

 

                DR. LAYLOFF:  I was going to comment on

 

      Art's question.  You know, if you demonstrate that

 

      your product complies and you cross-validate

 

      against that, then you can enter it in the columns

 

      because you would say it meets the specifications

 

      and it is up to someone else to demonstrate that it

 

      doesn't.

 

                DR. KIBBE:  Right.

 

                DR. LAYLOFF:  I mean, if you are

 

      challenged you don't have to retest it.  Their

                                                               174

 

      challenge has to include the test.

 

                DR. KIBBE:  Right, that is what I said.  I

 

      mean, as a manufacturer I don't have to continue to

 

      do this test at all.

 

                DR. LAYLOFF:  And if challenged you don't

 

      have to repeat it again.  They have to show that it

 

      doesn't comply.  The FDA has to go out and seize

 

      it.

 

                DR. COONEY:  Marv?

 

                DR. MEYER:  In one of your slides on page

 

      three you said inter-laboratory variability is a

 

      major contributor to the width of ranges.  If so, I

 

      wonder why that is the best way of doing it.  Why

 

      not just have a really good machine in a really

 

      good laboratory and do it?  FDA has a beautiful

 

      guidance on analytical methods for bio. studies and

 

      they specify variability, and they specify

 

      precision, and they don't say, well, this only

 

      applies to University of Tennessee but Pfizer has

                                                               175

 

      to do this and Teva has to do that.  They say this

 

      is what works; this is what we will accept.  Why

 

      not have a methodology that doesn't include inter;

 

      the inter is their problem it should be the USP

 

      problem.  You should just have a good set of data

 

      and everyone has to match it.

 

                DR. BROWN:  One of the things that you see

 

      in dissolution labs is that there are apparatuses

 

      that have tendencies.  I am not sure what

 

      constitutes a really good apparatus.  I am not sure

 

      that we have nailed it down even with the arbitrary

 

      limits that were given in Dr. Buhse's presentation

 

      this morning.  I am not sure what constitutes

 

      active variables in the dissolution.  I am not sure

 

      that any of us are.  So, I am not sure what that

 

      really good apparatus would be and would it reflect

 

      reasonable expectations from various

 

      well-manufactured products, apparatus products.

 

                DR. COONEY:  Tom?

 

                DR. LAYLOFF:  The dissolution experiment

 

      is not a mystical science enterprise.  Many years

 

      ago we did an 11-lab collaborative study of FDA

                                                               176

 

      labs where we required them to meet the system

 

      suitability requirement before they continued the

 

      test.  Now, what that paper said--and this was done

 

      I guess about 1990 maybe, what it said was that the

 

      between lab results variance was about 2.6 percent;

 

      the within lab was about 1.6, which is absolutely

 

      consistent with the retrospective study that was

 

      done by Bill Horowitz over about 50 studies in

 

      AOAC, collaborative studies among labs.  The among

 

      lab results for various analytical techniques for

 

      pharmaceuticals was 1.6 and the among lab data was

 

      about 2.6.  The ratio is about 1.7.  And the

 

      collaborative study for dissolution testing with 11

 

      FDA laboratories, all required to meet a

 

      suitability requirement, was in the same range,

 

      which meant that the sample preparation with the

 

      dissolution medium had the same variance as

 

      analysis with other techniques.

 

                DR. COONEY:  Ajaz?

 

                DR. HUSSAIN:  I think the challenge we

 

      face is if inter-lab variability is a concern,

 

      which I am not sure--and to some extent I am

                                                               177

 

      following what Tom is saying--I don't see how the

 

      calibrator really does anything.  It adds probably

 

      the majority of the variability to that study.  If

 

      we are going to make a calibrator using same type

 

      of excipients, raw materials with control of

 

      particle size based on D50 percent and so forth,

 

      how can that really give us any information when

 

      the quality of the product being tested--the real

 

      product might be far superior?  That is the

 

      fundamental flaw here.

 

                At the same time, what is the quality

 

      control strategy for the dissolution calibrator

 

      tablet?  It is another dissolution test.  So, that

 

      goes in circles.  So, that is the challenge we

 

      face.

 

                DR. COONEY:  Tom?

 

                DR. LAYLOFF:  One other thing, as was

 

      noted previously, the calibrator tablet isn't

 

      calibrator because you can't use it to calibrate

 

      anything.  It is a system suitability test tablet.

 

      As Ajaz mentioned, it in itself can be a moving

 

      target and your product may be less of a moving

                                                               178

 

      target.  If you have good control of your product,

 

      if you do have control over your particle size,

 

      excipient, polymorph--you control all those

 

      variables you are home free.

 

                DR. COONEY:  It seems to me we are coming

 

      back to a point that we talked about earlier, and

 

      that is to have clarity in what it is we want to do

 

      with the test that we are using, and to make sure

 

      that we are not just simply taking a methodology

 

      developed at some point in history and applying it

 

      to meet some regulatory or perceived regulatory

 

      requirement, and we need to really understand what

 

      we are testing for; what we want to measure; and

 

      what is driving the variance in those measurements.

 

                Are there any further comments from the

 

      committee?  Tom?

 

                DR. LAYLOFF:  One comment, it is a

 

      regulatory requirement and people lose product on

 

      the market because of it.  I mean, it is a very

 

      real thing that is out there.

 

                DR. COONEY:  Thank you very much.  This is

 

      the only presentation we have for the public

                                                               179

 

      period.  Since there are no others we can begin to

 

      go back to the regular agenda.  The next

 

      presentation, by Lawrence Yu, is on establishing

 

      drug release and dissolution specifications.

 

           Factors Impacting Drug Dissolution and Absorption:

 

                        Current State of Science

 

                DR. YU:  Good afternoon, everyone.  The

 

      assignment today is to talk about the factors

 

      impacting drug dissolution and absorption: current

 

      state of the science.  This certainly is a big

 

      title.  My talk will cover three aspects, basically

 

      in vitro dissolution testing. I am going to share

 

      with you some of the limits to oral drug absorption

 

      which has relevance to in vitro dissolution, and

 

      finally I want to share with you some thoughts on

 

      challenges to regulatory evaluation of dissolution

 

      which have been discussed this whole morning.

 

                This morning we discussed the variability

 

      of dissolution testing.  We discussed how to set

 

      specifications from a biopharmacist and

 

      pharmacokinetics perspective.  We discussed it from

 

      the manufacturing and control perspective.  The

                                                               180

 

      question is why are we doing dissolution testing?

 

      Why do we dissolution testing for almost all single

 

      solid oral dose drug forms, as well as the majority

 

      of dosage forms like parenteral dosage forms?

 

                Let's review the basic process of oral

 

      drug absorption when a patient takes a tablet or

 

      capsule.  This tablet or capsule will disintegrate

 

      or dissolve in the stomach.  Dissolved and

 

      undissolved drug will be emptied from the stomach

 

      into the small intestine where dissolution

 

      continues to occur.  The dissolved drug will cross

 

      intestinal membrane, will pass through the liver

 

      and reach the systematic circulation.

 

                So, from the mass transport perspective,

 

      look at the processes, the fundamental processes

 

      going on here.  We have gastric emptying, transit,

 

      dissolution, permeation, and metabolism.  These

 

      processes determine the rate and extent of

 

      absorption, all of which we call bioavailability.

 

                Because of the significance of

 

      bioavailability with respect to safety and efficacy

 

      of product, as you can see, dissolution becomes an

                                                               181

 

      essential or critical step, the first step for drug

 

      absorption.  That is part of the reason why.

 

      Another reason why we normally conduct dissolution

 

      test.

 

                Now, the question is what are we

 

      measuring?  Usually we use the classical equations.

 

      I understand those equations are not perfect.

 

      Sometimes they do not fit the dissolution profile

 

      well but they give you a flavor of what are

 

      critical variables involved with respect to

 

      dissolution.

 

                We have particle size which we mentioned

 

      this morning.  Basically, particle size effects the

 

      surface area, so surface area affects the

 

      dissolution.  The larger the surface area, the

 

      smaller the particle size, the faster the

 

      dissolution.

 

                We have solubility.  Now, a number of

 

      factors impact solubility, for example pH for

 

      ionized compounds.  For example, polymorphism

 

      impacts solubility.  So, those factors affect

 

      solubility which eventually affect dissolution. 

                                                               182

 

      That is why we say the particle size, polymorphism

 

      sometimes have impact on dissolution.

 

                Finally, there certainly is bulk media.

 

      Bulk media will determine the pH and pH, in turn,

 

      impacts the solubility and, again, impacts the

 

      dissolution.  Therefore, if we review the factors

 

      involved, we have particle size; we have solubility

 

      and certainly bulk solvent.

 

                In terms of utility, normally we have two

 

      utilities involved, two kinds.  Certainly under

 

      each kind there are different kinds of utilities.

 

      The first is for quality control from chemistry

 

      manufacturing control perspective.  I will give you

 

      an example here.  There are two polymorphic forms.

 

      The polymorphic II automatically translates into

 

      polymorphic I, for whatever reason such as

 

      manufacturing, storage, and so on and so forth.  It

 

      will impact dissolution.  So, therefore,

 

      dissolution is a tool for quality control.

 

                Another utility which we mentioned this

 

      morning is so-called in vivo performance

 

      evaluation.  Now, what I show you here is very

                                                               183

 

      beautiful, perfect examples.  I have to say we have

 

      a every chance to be successful in showing level A

 

      in vitro and in vivo correlation.  Nevertheless,

 

      this is a real example where one of the companies

 

      was able to come out with a beautiful IVIVC which

 

      certainly can be used for waiver in the future by

 

      current studies, which certainly can be used for

 

      setting specifications, and so on and so forth.

 

      But it is not easy.  It is a challenge to establish

 

      in vitro and in vivo correlation.

 

                Now, despite its utility for dissolution,

 

      there are many limitations.  When we talk about the

 

      limitations in the CMC or bio. area dissolution is

 

      always a hot topic because it is so easy to

 

      criticize.  For example, you will say the

 

      dissolution is over-discriminating, which means

 

      that in vitro dissolution you find a significant

 

      difference and you find no difference at all in

 

      vivo.  But in some cases you find no difference in

 

      in vitro dissolution, yet, you find a significant

 

      difference in vivo.  So, we call it

 

      over-discriminating.

 

                So, in order to understand those phenomena

 

      from the perspective of oral drug absorption, we

 

      have to understand the oral drug absorption first

                                                               184

 

      and particularly the limits to oral drug

 

      absorption.  That is what I want to talk about,

 

      limits to oral drug absorption.

 

                First let's review drug substance factors

 

      and then we will review the drug product factors,

 

      which is the disintegration.  When we review drug

 

      substance we mentioned that particle size and

 

      solubility are two major factors which impact the

 

      in vitro absorption and certainly sometimes, again,

 

      in vivo absorption.  So, in order for us to

 

      understand the limits we have to define what are

 

      the limits here.  Yes, we have dissolution but in

 

      vivo absorption certainly involves more than just

 

      dissolution, as we discussed.  In terms of process,

 

      we have transit, gastric emptying.  We have

 

      permeation and metabolism.

 

                But from an in vitro perspective, from in

 

      vivo dissolution perspective what we define as the

 

      limits to oral drug absorption is dissolution

                                                               185

 

      limited absorption and solubility limited

 

      absorption and permeability limited absorption.

 

      Now, it is very easy to understand what is called

 

      permeability limited absorption.  It is simply that

 

      the drug across membranes is very, very slow

 

      compared to other process.  That is why it is

 

      called limited.

 

                Now, with dissolution limited absorption

 

      we have to talk about dissolution so the

 

      dissolution limited absorption seems very easy to

 

      understand.  People ask me very often what is

 

      called solubility limited absorption.  Why do you

 

      distinguish solubility limited absorption from

 

      dissolution limited absorption?  This is because in

 

      the reality setting, especially for drug discovery

 

      and development, we have to understand that the

 

      concentration in vivo or in the gastrointestinal

 

      tract of a patient cannot exceed the solubility, as

 

      is shown here in the middle.  Concentration cannot

 

      exceed solubility.  Now, with in vitro dissolution

 

      testing we always have simulated conditions so,

 

      regardless of the amount of drug, it can always be

                                                               186

 

      dissolved but not necessarily in vivo.  Think about

 

      it, if you dump tons of drug into the human body,

 

      they are totally saturated and the concentration

 

      cannot exceed solubility regardless of how much you

 

      dump into the patient or subject.  It is not quite

 

      useful anymore.  This is the distinguishing

 

      difference between solubility limited absorption

 

      and dissolution limited absorption.

 

                Mathematically, with dissolution limited

 

      absorption we generally refer to particle size as a

 

      major factor.  Solubility limited absorption refers

 

      to solubility.  Now, solubility, of course, in turn

 

      impacts dissolution but from an in vivo

 

      perspective, because the concentration cannot

 

      exceed solubility, that is why we define solubility

 

      limited absorption.  It has clinical implications

 

      with respect to permeability limited absorption,

 

      solubility limited absorption and dissolution

 

      limited absorption.  Let's look at it.

 

                Theoretically, in order for us to define

 

      under what conditions is dissolution limited, under

 

      what conditions is solubility limited we have

                                                               187

 

      designed some numbers and we have dose volume; we

 

      have dissolution time; we have absorbable dose.

 

      The dose volume is the amount required to dissolve

 

      the dose.  The dissolution time is minimal time

 

      required to dissolve a single particle under the

 

      same conditions, while absorbable dose is the

 

      maximum amount of drug that can be absorbed under

 

      certain physiological conditions such as solubility

 

      and transit time.

 

                Based on those parameters, we can define

 

      under what conditions is dissolution limited; under

 

      what conditions is permeability limited; and under

 

      what conditions is solubility limited.

 

                Let's look at the comment here.  For

 

      dissolution limited absorption or for permeability

 

      limited absorption the absolute amount of drug

 

      increases with the increased dose.  So, it is very

 

      simple.  When you require high exposure in vivo

 

      simply give more drug.  That is usually the

 

      practice in drug discovery and probably

 

      development, particularly during animal toxicity

 

      studies.

 

                However, for solubility limited absorption

 

      the absolute amount of drug absorbed does not

 

      increase with increase of dosing.  That is part of

                                                               188

 

      the reason that for solubility limited absorption

 

      you have to rely on other approaches to improve

 

      bioavailability, to improve the absolute amount of

 

      drug absorbed.  Very often industry and our fellow

 

      friends in toxicology department they often give

 

      more and more drug and when they cannot see high

 

      exposures they ask us why.  The reason is that in

 

      many cases it is because the absorption becomes

 

      solubility limited, therefore, if you give more and

 

      more drug you cannot see absolute increase of the

 

      amount absorbed.

 

                In terms of prediction--and this is the

 

      permeability limited absorption, there is certainly

 

      fraction of dose absorbed, although percent of drug

 

      absorbed can be reasonable predicted with

 

      mathematical equations.  The data in this slide is

 

      from human permeability but sometimes rat

 

      permeability can be used to give us very good

 

      results.  K2 cell permeability can be used but does

                                                               189

 

      not necessarily give us good results, in other

 

      words, whether in humans the compound has high

 

      permeability, can always high permeability in other

 

      systems, including the rat.

 

                This is another beautiful example to

 

      determine how particle size impacts absorption.  As

 

      I mentioned, in this theoretical framework we

 

      define dissolution limited absorption is because of

 

      particle size so, as you can see, for digoxin, yes,

 

      it is very poorly soluble; yes, the solubility is

 

      only 60 mcg/ml.  You are actually able to get 100

 

      percent bioavailability.  In this case there is no

 

      metabolism, therefore, 100 percent absorption

 

      equivalent to the bioavailability.

 

                This morning we discussed particle size

 

      distribution, and I have to tell you that in

 

      simulation in these slides I simply used mean

 

      particle size.  Yes, there is literature out there

 

      using particle size distribution in order to

 

      understand how particles impact absorption.  In

 

      fact, in my publication we attribute that at 100

 

      mcg prediction is not that good because we used

                                                               190

 

      mean particle size instead of particle size

 

      distribution because even though mean particle is

 

      100 mcg, a number of small particles can be full

 

      absorbed.  That is part of the reason that you see

 

      higher percent absorption, much higher than what

 

      has been predicted.

 

                Griseofulvin is a very classical example.

 

      If you look at 100 mg griseofulvin, the solubility

 

      is 95 mcg/ml.  As you can see, when you give a

 

      higher dose the fraction of dose absorbed has not

 

      been increased, and part of the reason is when you

 

      reduce particle size.  It explains the utility of

 

      solubility limited absorption.

 

                This is a graph never published but I

 

      thought I would share it with you because it is the

 

      interplay of bioavailability, solubility,

 

      permeability and hepatic clearance, showing in the

 

      drug discovery setting of the drug is above the

 

      surface.  This means the bioavailability most

 

      likely is lower than 30 percent.  Below this, the

 

      bioavailability is most likely above 30 percent.

 

      So, if you want to increase the bioavailability,

                                                               191

 

      depending on where the drug molecule is located,

 

      you can increase the solubility; you can increase

 

      the permeability; you can increase hepatic

 

      clearance and increase the bioavailability.  So,

 

      that is the utility in industry settings.

 

                This is a very brief overview of how

 

      factors impact the absorption from the drug

 

      substance perspective.  This will give a very brief

 

      overview about the drug product factors which

 

      impact the absorption.  Certainly, the

 

      manufacturing process could impact the particle

 

      size but I will mainly focus on disintegration, in

 

      other words, how a tablet disintegrates and becomes

 

      smaller particles.

 

                In order to illustrate how the

 

      manufacturing process could impact dissolution, I

 

      will just give you an example here.  This is a drug

 

      which is highly soluble and highly permeable.  So,

 

      if you give oral solutions you can see the Tmax at

 

      about 1.3 hours and it very quickly reaches the

 

      Cmax, about 13 hours.  This is basically the

 

      solution curve.

 

                Now, when you give a different dosage

 

      form, because of slowing of disintegration you

 

      could have a different dissolution profile, slow,

                                                               192

 

      medium and fast.  When we translate this in vitro

 

      dissolution into in vivo here is the plasma

 

      concentration profile.  Obviously, the slower the

 

      solution translates into lower Cmax and lower AUC.

 

                Another case is when you have a different

 

      coating system, this may be dissolved at different

 

      pHs and in in vitro dissolution we are able to see

 

      a significant difference and you also see it in

 

      vitro, as you can see in this slide.

 

                So, those slides basically illustrate how

 

      the dosage form impacts dissolution, impacts

 

      absorption.  Certainly, there are many factors in

 

      in vitro dissolution that impact, for example,

 

      manufacturing process, compression force, and so on

 

      and so forth.  Those will impact the dissolution of

 

      a product.

 

                I want to share with you some thoughts

 

      about challenges or opportunities which we are

 

      facing today.  First of all, when dissolution is

                                                               193

 

      very rapid, outcome rate determining steps, drug

 

      levels in the blood and plasma may not reflect

 

      dissolution differences at all, as you can easily

 

      understand.  Because dissolution is not limited in

 

      vivo, therefore, in vitro how much different they

 

      are doesn't matter because you cannot see it in

 

      vivo from the in vivo perspective.  Yet, for

 

      dissolution limited absorption sometimes we can get

 

      IVIVC and sometimes actually in vivo fraction

 

      absorbed is not always possible because of lack of

 

      IV.

 

                For solubility limited absorption, as well

 

      as permeability limited absorption in vitro

 

      dissolution will not always reflect into in vivo

 

      because they are limited by solubility, because

 

      they are limited by permeability, therefore, any

 

      difference in vitro may not be translated into the

 

      in vivo at all.  So, we normally call that

 

      over-discriminating.

 

                Now, here we have f2, very useful,

 

      similarity factors.  Usually we use 50 or above

 

      because 50 or above reflects average 10 percent

                                                               194

 

      difference or less.  I have to say this f2 value

 

      certainly works fine or works well in most cases

 

      but we can always see a criticism here because in

 

      some cases the f2 value is not reflected in vivo

 

      because, as I said, the f2 value is basically

 

      average 10 percent difference.  When you average 10

 

      percent under different limits to absorption, those

 

      may be reflecting in vivo or may not--may not see

 

      in vivo situation.

 

                So, as I said earlier in the discussion of

 

      the presentation, there is a role of dissolution

 

      for quality control and for in vivo performance

 

      evaluation.  So, the question that comes to my mind

 

      is are these two goals always consistent?  They may

 

      not be.  That presents an opportunity for us.  For

 

      example, under the hydrodynamic conditions--and we

 

      have a paper from Rutgers University to show that

 

      Reynolds number for chemical engineering for 50 rpm

 

      as well as for 100 rpm--under those conditions we

 

      may see some in vivo/in vitro correlations.  In

 

      fact, the paper suggests we may lower the rate so

 

      that under laminar flow we could see significant or

                                                               195

 

      more difference in the amount of dissolution.  Yet,

 

      when we go to the lower levels number and in

 

      laminar flow, even though we may see in vitro, but

 

      those may not translate in vivo because in vivo the

 

      hydrodynamics is much more higher, the Reynolds

 

      numbers--could be a lot higher.   In fact, in the

 

      stomach the motility is highest--educated guess, it

 

      is difficult to see in vitro laminar flow in the

 

      same situation.

 

                And then there is the media.  The media is

 

      always an issue because in the real setting we will

 

      evaluate a number of the media and we are trying to

 

      define the dissolution a significant difference, or

 

      most of the difference when we select in vitro

 

      dissolution testing.  If you make a survey of some

 

      of poorly soluble drugs, for example, sometimes we

 

      saw hydrochloride.  Sometimes we see pH 7.4

 

      phosphate buffer--these are all dissolution tests.

 

      The question that comes to us is are those

 

      dissolution tests or dissolution media reflected in

 

      vivo?  I have serious doubt that our human

 

      beings--sometimes the pH could be 7.8 in the

                                                               196

 

      stomach or sometimes could be 7.4, or sometimes the

 

      concentration could be 2 percent or sometimes could

 

      be zero percent.  I think there is a wide spectrum

 

      maybe in vivo.  This is true but it may not change

 

      as much as we see in in vitro dissolution

 

      evaluation.

 

                So the question that comes back is should

 

      these two objectives for dissolution testing be

 

      separated?  I think this is the challenge in front

 

      of us.  We do not have a solution yet but I think

 

      we ought to seriously look at the possibility and

 

      the value and drawbacks as well as benefits.

 

                For example, for dissolution quality

 

      control, hopefully, hydrodynamics and media are

 

      chosen for reproducibility and detection of product

 

      changes, for example, particle size changes; for

 

      example, polymorphic changes.  Certainly, design of

 

      in vitro dissolution test and for quality control

 

      are not constrained by a desire to mimic in vivo

 

      conditions.

 

                But for in vivo biorelevant dissolution we

 

      may choose a battery, a number of tests which

                                                               197

 

      pretty much covers what is going on in vivo in the

 

      human body.  So, those tests will, hopefully, get

 

      reasonably good correlations to in vivo.  Yes, FDA

 

      has lots and lots of dissolution data.  Yes, we

 

      have a lot of products approved that have required

 

      dissolution data.  Yet, when we look at dissolution

 

      data and try to transfer those dissolution data

 

      into knowledge, unfortunately, we almost get

 

      nothing because every single drug, every single

 

      product has used similar or even different

 

      dissolution media.  It has been difficult for us to

 

      get to some kind of in vivo/in vitro correlation

 

      even though we have lots, and lots, and lots of

 

      data because the difference among in vitro

 

      dissolution tests almost cannot be translated in

 

      vivo.  That is the difficulty.

 

                So, in summary, believe me, I have

 

      discussed in vitro dissolution testing and

 

      discussed the limits to oral drug absorption.

 

      Again, the limits to absorption could be

 

      solubility; could be dissolution; and could be

 

      permeation.  We have briefly overviewed the

                                                               198

 

      dissolution profile comparison and discussed the

 

      future role of dissolution with respect to quality

 

      control and in vivo performance evaluation.  Thank

 

      you very much.

 

                     Questions by Committee Members

 

                DR. COONEY:  Thank you.  I would now like

 

      to invite questions from the committee.  Ken?

 

                DR. MORRIS:  I guess, Lawrence, one thing

 

      that occurs to me is that really the distinction

 

      between your dissolution and solubility limitation

 

      seems to me--or, I guess this is a question, are

 

      you really differentiating the source of the

 

      solubility attenuation?  In other words, aren't you

 

      really looking at whether or not it is the activity

 

      coefficient or the lattice energy that is

 

      controlling the solubility?

 

                DR. YU:  I guess when we talk about

 

      solubility, certainly there are two parameters.  In

 

      this difference, when we distinguish the solubility

 

      limited absorption and dissolution limited

 

      absorption, we infer that solubility is the

 

      solubility final number, whether it is caused by

                                                               199

 

      lattice energy or not.

 

                DR. MORRIS:  But that is not really the

 

      case, is it?  Right?  I mean, there is a

 

      distinction.  I think it only changes the

 

      terminology; I don't think it changes your

 

      conclusions but maybe we can talk about it

 

      off-line.

 

                DR. KIBBE:  I follow him a lot better than

 

      where you are going.

 

                [Laughter]

 

                DR. YU:  Thank you, Art.

 

                DR. KIBBE:  What he is talking about is

 

      absorption and what drives absorption, and not

 

      dissolution.  What drives dissolution is a

 

      different set of parameters.

 

                DR. MORRIS:  I don't think so.  If you are

 

      talking about the absolute value--I mean, you have

 

      the same solubility but different causes for--

 

                DR. KIBBE:  No, he is talking about

 

      different absorption with different solubility.

 

                DR. MORRIS:  But that gives you the

 

      differences for the driving force.  The driving

                                                               200

 

      force is determined by the concentration or the

 

      activity--

 

                DR. KIBBE:  Right.

 

                DR. MORRIS:  --no matter what.

 

                DR. KIBBE:  What he is saying is that you

 

      have a series of events and if the rate of

 

      dissolution gets you to a relatively low solubility

 

      maximum quickly, then the whole thing is driven by

 

      the limited solubility in terms of absorption.  We

 

      can do the numbers off-line.

 

                DR. MORRIS:  Yes.  Yes, I see what you are

 

      saying.  I still think that your dissolution is

 

      solubility limited.  It is still just a different

 

      solubility limitation, but we can talk off-line.

 

                DR. COONEY:  Ken, I think the point you

 

      are making relates to one segment of the possible

 

      space where you are solubility limited.

 

                DR. MORRIS:  Yes, for low solubility.

 

                DR. COONEY:  Yes.

 

                DR. YU:  I guess the solubility limited

 

      absorption as well as dissolution limited

 

      absorption is with respect to in vivo absorption. 

                                                               201

 

      You have to look at in vivo absorption when we talk

 

      about those limits because with in vitro

 

      dissolution evaluation you always same conditions,

 

      at least based on FDA guidance.  Therefore, you

 

      always can see dissolution.  But in vivo there is

 

      dissolution going on but think about when the whole

 

      small intestine is saturated by the drug, under

 

      this condition we call it solubility limited

 

      absorption.  I don't know if I can explain it to

 

      you better.

 

                DR. MORRIS:  I think I understand what you

 

      are saying.

 

                DR. YU:  It is just terminology I guess.

 

                DR. COONEY:  Marvin?

 

                DR. MEYER:  Lawrence, the dissolution for

 

      quality control is distinct from dissolution for in

 

      vivo, that comparison.  Why have two?  If your

 

      quality control is irrelevant to in vivo, I don't

 

      see any particular relevance to it.  Let's say you

 

      want to have something you can detect changes in

 

      the product, but what if you needed 25 percent

 

      methanol in water to detect a change in coating

                                                               202

 

      thickness, and you needed 10 percent methanol in

 

      water to detect a change in particle size, and you

 

      needed some other medium to detect a change in the

 

      combination of the two and then, all of a sudden,

 

      you would have three or four dissolution tests,

 

      none of which may make any difference when it comes

 

      down to the various product differences in the in

 

      vivo setting?  Why hot just focus in on the

 

      biorelevant dissolution?

 

                DR. YU:  Well, I think the question that

 

      comes back is that, first of all, in the 40 years

 

      of dissolution history it is quite unusual.  As I

 

      mentioned, you really need a lot of laughing,

 

      sunshine and good luck to get in vivo/in vitro

 

      correlation, and even if you get it today it may

 

      not exist tomorrow if you change the formulation a

 

      little bit.  That is why the famous words from Ajaz

 

      are that IVIVC is formulation specific.

 

                The question is why do we need to

 

      separate?  What is the value?  I guess this was

 

      presented to you today.  What I would argue is the

 

      following, when we talk about in vivo dissolution

                                                               203

 

      testing it is quite complex.  It is not so

 

      difficult to do.  If you think about it, you have

 

      100 products, or one company has, you use 900

 

      vessels and each and every day you consume a lot of

 

      acid.  So, if you think about it, those dissolution

 

      tests can be replaced by simple quality control

 

      dissolution tests of, say, water it is a worthwhile

 

      effort simply from an economic perspective and from

 

      a convenience perspective.  However, you have to

 

      say whether those tests are sensitive enough to

 

      detect any significant changes in vitro, for

 

      example particle size changes, polymorphic changes,

 

      and so on and so forth.  But in vivo dissolution

 

      tests, hopefully, is a fixed battery of dissolution

 

      tests which pretty much capture what is going on in

 

      the physiological conditions.  Now, whether we

 

      actually can develop those or not we are waiting to

 

      see.  This is simply a proposal presented to you to

 

      see what you think about it.

 

                DR. HUSSAIN:  May I?  I think just to add

 

      to what Lawrence just said, Marvin, the current

 

      tendency is to use--the phrase "performance test"

                                                               204

 

      was used to some extent--is to capture all the

 

      other controls that are missing.  Let me explain

 

      that.  We currently have a univariate focus on

 

      quality.  What it means is we do all the testing on

 

      the content, rate and everything of the drug

 

      substance only.  But the excipients, distribution,

 

      and so forth, is also relevant.  So, dissolution

 

      tests for quality purposes capture all those

 

      aspects.  Actually, I have an example in my slides

 

      that I could show you later on.

 

                So, that is the current philosophy of

 

      needing a dissolution test even if it is not

 

      relevant from an in vivo perspective to do that.  I

 

      think quality by design would say that if you are

 

      controlling all aspects that are relevant, then you

 

      may not even need one.  I mean, we opened that

 

      possibility to you.

 

                DR. COONEY:  Art?

 

                DR. KIBBE:  Which is kind of where I

 

      wanted to go.  The dissolution testing and a lot of

 

      our terminal testing was developed when we couldn't

 

      characterize our product effectively.  The easiest

                                                               205

 

      way of determining whether your tablet was hard

 

      enough was to snap it and listen to the sound of it

 

      cracking it.  Friability was you took a handful and

 

      dropped them on the floor, and if none of them

 

      broke then your tablet was okay.  That is where I

 

      started in this business so, hopefully, we have

 

      come a long way from that.

 

                If we have a process under control that we

 

      can characterize and we know the factors that are

 

      affecting the way it performs in vivo because we

 

      have looked at them with data in people, then you

 

      say to yourself do I still have to have a test

 

      which was originally thought of as a surrogate for

 

      the in vivo performance at all?  If so, what is it

 

      getting me in terms of information to help control

 

      my process?

 

                The question I asked USP was, well, can we

 

      still be USP without doing this test?  Of course,

 

      you can.  You don't need to do this test if you

 

      will eventually pass it whenever you felt like

 

      doing it.  You could do tests that are much more

 

      useful and instructive in terms of whether your

                                                               206

 

      process is under control.  It reminds me of the law

 

      that is still on the books in New York City that

 

      when the car was invented the requirement was that

 

      a man walk 20 paces in front of the car with a red

 

      flag to warn everybody that the car was coming.

 

      Now if they tried to enforce that law in New York

 

      he would get run down by the 40 cars that were

 

      coming.  Perhaps we ought to look at dissolution

 

      testing as the guy with the red flag, warning us

 

      that we might not be in control anymore when we

 

      really have much better controls on the system.

 

                DR. COONEY:  Are you suggesting we change

 

      the red to a blue flag?

 

                [Laughter]

 

                DR. KIBBE:  I was going to go for Claude

 

      Raines and make the flag invisible.

 

                DR. MORRIS:  Actually, it is Kevin Bacon

 

      these days.

 

                DR. KIBBE:  That shows you my age, right!

 

                DR. MORRIS:  I tried a Claude Raines in

 

      class and nobody knew what I was talking about.  I

 

      guess to that point, if you can characterize it, I

                                                               207

 

      mean sort of looking at statistical significance

 

      without getting into discussion of statistics, you

 

      can have a much larger fraction of the population

 

      sampled during development than you are going to

 

      ever have when you are doing manufacturing.  So,

 

      once you establish that design space, then by the

 

      time you get to manufacturing, in principle you

 

      should never have to do dissolution, which I am

 

      sure would make everybody a lot happier if not a

 

      little nervous.

 

                DR. COONEY:  Tom?

 

                DR. LAYLOFF:  I can see dissolution

 

      testing as a regulatory tool to assure that the

 

      manufacturer is putting out a product which

 

      conforms to the specifications, but there is no

 

      recovery from a failure in dissolution tests, short

 

      of destroying the batch.  I think that there have

 

      been in place for a very long period of time the

 

      tools to control the processes so that you can

 

      deliver a product which would meet the test if

 

      tested.  So, I don't see the dissolution test as a

 

      useful release test because if it fails that

                                                               208

 

      release test you have failed the lot.  It means the

 

      process has failed, everything has failed.  So, I

 

      don't see it as a utility for a release test.  I

 

      see it as a regulatory tool, enforcement tool

 

      because you can see using that to hammer somebody

 

      but I can't see it as a release tool.  If you have

 

      to use dissolution testing as a release tool it

 

      means you don't have everything in control.

 

                DR. COONEY:  I think what we will do is to

 

      move forward to the next presentation.  There is

 

      clearly going to be more conversation and

 

      discussion and that will be most appropriate after

 

      Ajaz has taken us through his first 16 slides.

 

                [Laughter]

 

                        Summary of Tactical Plan

 

                DR. HUSSAIN:  Yes, I will be brief and

 

      just lay out the steps of the tactical plan and

 

      stop at that point and open this for discussion.  I

 

      do have a number of slides in your handout which go

 

      on to sort of illustrate some more deeper part what

 

      we are thinking about and some illustrative

 

      examples.

 

                But just to summarize, I think what we are

 

      trying to do is seek your recommendation on are our

 

      tactical steps outline consistent with the goals we

                                                               209

 

      are trying to achieve?  What additional steps, if

 

      any, of changes would you recommend to improve our

 

      plan?  What additional scientific evidence is

 

      necessary to support development of the plan when

 

      we come back to you to make the proposal next time,

 

      hopefully?  General considerations for identifying

 

      and developing statistical procedures; any other

 

      specific recommendations you may have.

 

                Now, the proposed steps are in one clump

 

      focusing on the measurement system, an alternate

 

      regulatory approach, suitability of dissolution

 

      measurement system which will rely on a rigorous

 

      mechanical calibration and, when necessary,

 

      measurement of degassing concepts that Cindy talked

 

      about.  This will be coupled with a

 

      characterization of your clinical pivotal lot or

 

      the bio. lot in terms of a gauge

 

      reproducibility/repeatability study where you could

 

      look at how sensitive this formulation is to

                                                               210

 

      conditions of measurement systems such as

 

      degassing, such as operator, such as the apparatus,

 

      and so forth, in a structured design in an

 

      experimental way since this is a destructive

 

      sample, and there are means to do that.

 

                Then we will focus on developing decision

 

      trees, and I have put them in one clump.  The focus

 

      of the decision tree would be to establish

 

      dissolution rate specification.  I have requested

 

      Lawrence to give you a flavor, just a flavor of how

 

      we can start thinking from a mechanistic

 

      perspective of setting mechanism-based

 

      specification.  He just gave you a snapshot of the

 

      mechanisms that affect absorption.  There is a

 

      whole other set of mechanisms that define the

 

      release of drug from the dosage forms, and so

 

      forth, and depending on the types of release

 

      mechanisms so that combination will allow us to

 

      start thinking about how you approach a

 

      mechanistic-based specification because that will

 

      drive us to what are the critical factors that

 

      affect release, and then work around that.

 

                This is also an opportunity because often

 

      people are concerned that when you talk about

 

      mechanism this will restrict--that only few

                                                               211

 

      mechanisms are acceptable.  No, I think it actually

 

      says you can have multiple different mechanisms but

 

      then you will modify your specifications to that

 

      specific mechanism and not force companies to one

 

      set of dissolution specifications.

 

                Clearly, I think the opportunities for

 

      utilizing a control philosophy, quality by design

 

      and our technologies under the umbrella of PAT will

 

      replace these methodologies.  As I mentioned, the

 

      methodology would be part of the decision tree.  As

 

      part of the decision tree also would be the level

 

      of process understanding and control to achieve--

 

      to essentially create a concept of design space and

 

      how this might be used for post-approval changes,

 

      the type of changes that Mehul sort of illustrated

 

      to you, and I will actually pick that up in my talk

 

      tomorrow and explain that further.

 

                Also, I think it is important for us to

 

      make sure our decision trees are compatible and

                                                               212

 

      equally open and transparent to all.  Therefore, we

 

      would like to develop a side-by-side comparison of

 

      our decision tree for new and generic drugs.  I

 

      think one of the key aspects would be to come to

 

      the committee to get an endorsement for the level

 

      of quality assurance and quality control confidence

 

      that we will have with our decision trees and our

 

      control strategy, quality by design, would be

 

      higher than what the current system is.  To me, it

 

      is a given but I think all of us need to be

 

      convinced of that.

 

                Today we will specifically seek

 

      recommendations from you on the statistical

 

      procedures, how do we want to proceed, and I have a

 

      couple of slides on that.  After this meeting we

 

      will get busy and develop a detailed proposal for a

 

      subsequent meeting for discussion.  The timing of

 

      this meeting was very important.  We rushed and

 

      tried to get this meeting because, as I said, this

 

      weekend I go to Brussels and we are starting a

 

      discussion on decision trees for dosage forms in

 

      ICH and I needed this discussion behind me, and the

                                                               213

 

      comfort of knowing, arguing and making the point

 

      because we want to achieve harmonization in Europe,

 

      Japan, and so forth.  So, this meeting and your

 

      recommendations I think will play a part in our

 

      discussions.

 

                Just to summarize what we intend to

 

      accomplish with all this--improve our ability to

 

      identify sources and type of variability, and to

 

      ensure quality by design.  Vibhakar illustrated one

 

      simple example to you.  If you only rely on

 

      dissolution, even though you have IVIVC, a

 

      correlation may not be causal.  I think you have to

 

      bring a control system perspective or control

 

      strategy to assure quality, not just a test.

 

                Obtain global estimates of variability to

 

      use in regulatory decisions.  Our current approach

 

      can be improved.  You saw our approach and

 

      opportunities for improvement there.  And, we would

 

      like to use this information on variability,

 

      sources of variability in how we set regulatory

 

      specifications and process controls so that we

 

      focus on controlling the real source of variability

                                                               214

 

      and, for example, if the concern is that this

 

      excipient is not uniformly distributed and that is

 

      important, today the only test we have is

 

      dissolution.  If you have other means you can move

 

      in that direction.  We also want to use this

 

      information for assessment of adequacy of proposed

 

      material and manufacturing process control

 

      strategies.

 

                Facility, assessment and communication of

 

      technology knowledge transfer and assurance of

 

      state of control and production operation--this is

 

      the current big gap between CMC and GMP, and I

 

      think this will help us to bridge that gap.

 

      Clearly, the basic philosophy is if you can

 

      demonstrate a state of control that opens the door

 

      for continuous improvement flexibility.

 

                The inspiration for the proposal--there is

 

      not a single thing which is new or unique in this

 

      proposal.  It is well established and we have

 

      simply borrowed it from other sectors.  The

 

      inspiration was the DMAIC concept of defined

 

      measure analyzing improvement control of the Six

                                                               215

 

      Sigma concept.  So, what we are proposing is in

 

      practice in every other sector, literally.

 

                But there are many challenges, and this is

 

      an important challenge that I think we have

 

      discussed several times, the pharmaceutical

 

      quality, because of the challenges--the consumer,

 

      the patients or the physicians cannot judge

 

      quality--creates some challenges.  Now, the key

 

      concepts in Six Sigma are that you need to know

 

      what is critical to quality attributes, attributes

 

      most important to the customer.  This is the

 

      quality to clinical challenge.  It affects failing

 

      to deliver what the customer wants.  In this case,

 

      failing to meet your specifications or deviations

 

      in GMP practices unless these really are focused on

 

      critical to quality attributes, and so forth.  This

 

      is an opportunity.  It is in green.  We can get to

 

      more critical variables and focus on signs rather

 

      than following what our practices have been.

 

                Process capability, what your process can

 

      deliver, this is again an opportunity because the

 

      concern from a CMC review perspective is that

                                                               216

 

      process validation may not be adequate, and with

 

      the sample size that we have from a USP market

 

      standard perspective as a release test is a

 

      significant concern, whether it is right or not.  I

 

      think that is debatable.  But the market standards

 

      are perfectly find.  In my opinion, I think the USP

 

      market standard is fine.  There is nothing wrong

 

      with that.  I think the practice of using them as

 

      release tests and as in-process tests, that is

 

      where the challenge is.

 

                But the challenge I think is variation,

 

      what the customer sees and feels.  We don't know

 

      this so we have to go with what our signs say and

 

      what the best practices are.  Dr. Woodcock, in a

 

      paper, called it market failure because you cannot

 

      get the feedback from the customer really unless

 

      there is a dramatic failure.

 

                Stable operation is an opportunity, and

 

      this is a significant opportunity for continuous

 

      improvement because the regulatory agencies around

 

      the world and industries also--corrective actions,

 

      the only leverage for continuous improvement--we

                                                               217

 

      have to get away from that mentality.  And,

 

      demonstration of stable operations can provide a

 

      logical and scientifically rigorous way to

 

      alleviate this concern.

 

                Design for Six Sigma I think is designing

 

      to meet customer needs and process capability, and

 

      here the fundamental premise is that you design

 

      your product for your patient, not for your

 

      dissolution test.  I mean, that distinction really

 

      has to come through.  And, many times I think if

 

      FDA is asking the right question we can make sure

 

      that happens.  If FDA doesn't ask the right

 

      question they will design to what FDA wants, and I

 

      think that is a fundamental challenge.  In some

 

      ways that is a specifications capability gap that

 

      can exist.  So, here are the opportunities and

 

      challenges and I think we have to address those as

 

      we develop our decision trees.

 

                Step number one is measurement system

 

      suitability, and this is clearly honing down on the

 

      target value, the mean value for your measurement.

 

      It focuses on mechanical and media factors, but I

                                                               218

 

      am not sure it is actually an independent step so I

 

      have also put step number two, gauge R&R which is

 

      essentially a qualifying one that you do once as

 

      part of your approval, using a clinical pivotal lot

 

      or a bio. lot.  Here you have to think about

 

      analysis of variance and the factors that might

 

      contribute, like apparatus, dissolution media,

 

      operator, clinical pivotal lot, and this a

 

      structure design of experiment.

 

                Now, clinical pivotal lot or gauge R&R has

 

      some considerations.  It is not just automatic.  It

 

      has to be supported by pharmaceutical development,

 

      stability and sampling.  By stability, here I mean

 

      the process was stable from start to finish, not

 

      from the conventional stability perspective also.

 

      It is a statistical control.

 

                Can we also do this for currently marketed

 

      products is a question mark.  Information that

 

      would come from these studies would help us

 

      facilitate a shift from a deterministic design

 

      culture because we want to move towards assessing

 

      variability and using variability.  So, this is

                                                               219

 

      step two.  This could be an independent step but I

 

      just wanted to sort of repeat that here.

 

                The decision trees--I am not going to walk

 

      you through in detail of the decision trees.  I do

 

      have some examples later on.  How should we

 

      consider moving towards decision trees?  Lawrence

 

      and the Office of Generic Drugs has started working

 

      on a question-based review process, and the whole

 

      thing is asking the right questions in a sequence

 

      that drives you towards quality by design.  So,

 

      clearly, as part of the decision trees is what are

 

      the key questions we should be asking, and so

 

      forth.  So, asking the right questions would be a

 

      consideration.  Clearly, beginning with the end in

 

      mind, which is the intended use, keep the focus on

 

      intended use as we develop these questions.

 

                Systems base, connecting the key

 

      disciplines and regulatory submission section--at

 

      the previous advisory committee, as part of the OPS

 

      Critical Path Initiative, I presented a proposal on

 

      how to connect different parts of the section that

 

      will be part of this consideration.  Vince Lee

                                                               220

 

      actually has started working on that and I think we

 

      will see how that connects.

 

                We also wish to facilitated structured

 

      product development process.  The traditional trial

 

      and error of one experiment at a time will not

 

      really cut it.  You really need to have a

 

      structured product development.  Yet, we do not

 

      want wish to dictate the specific process.  That is

 

      the challenge.

 

                Pre-approval changes and bridging

 

      studies--on average in a new drug application there

 

      are three to six bioequivalence studies done on new

 

      drugs--three to six.  Some we don't even review.

 

      If you really look at it, if you leverage that

 

      information to bring in considerations that every

 

      experiment you do is a hypothesis you have so many

 

      opportunities to evaluate that hypothesis.  So,

 

      bridging studies would be a leverage.

 

                The decision trees would be cumulative in

 

      terms of leading to a decision but also support use

 

      of prior knowledge.  For example, we have wonderful

 

      approaches to predict the impact of particle size,

                                                               221

 

      and so forth.  So, as you are in your early part of

 

      development studies you can predict the impact of

 

      particle size on manufacturability as well as

 

      dissolution and bioavailability.  So, if you have

 

      such a system in place for the next product that

 

      comes along you will make the prediction and

 

      evaluate that so you postulate your next experiment

 

      that you go into in vivo that this is what I

 

      expect.  Once you do that, you start setting up a

 

      learning system that at some point becomes very

 

      useful.

 

                That means that we will move towards a

 

      scientific hypothesis format.  I have removed those

 

      slides.  Those slides come later on if you have any

 

      questions.

 

                How do se address the challenge, the two

 

      challenges of market failure and quality to

 

      clinical gap?  In a sense, one of the challenges I

 

      think we have is we think about risk and

 

      uncertainty and we confuse that, and this is

 

      again--I learned from this committee and I will

 

      share with you a table of how we can separate

                                                               222

 

      uncertainty, variability and risk tomorrow.  I

 

      shared it at the training session yesterday with

 

      some of you.

 

                I think our current assumption that we

 

      work under is that our methods that we use are most

 

      discriminating, therefore, risk is mitigated.  So,

 

      if that is the case we are not dealing with risk;

 

      we are dealing with uncertainty and how you

 

      approach that opens up a whole new set of

 

      approaches.  So, if you are dealing with

 

      uncertainty lack of knowledge is the challenge.

 

      So, improve the knowledge that leads you to that.

 

      So, from that basis, product specification based on

 

      mechanistic understanding provides a means to

 

      address uncertainty and that will be a progression

 

      that we will use.

 

                So, if you really look at it, the way we

 

      set specifications is because of uncertainty.

 

      Uncertainty management without pharmaceutical

 

      development information is a challenge.  We focus

 

      on a discriminating test concerned with in vivo but

 

      without the pharmaceutical development information.

                                                               223

 

      The discriminating part comes from the method

 

      measurement system.  Is it really discriminating

 

      the formulation variables that really impact?

 

      Sometimes you have a disconnect there.

 

                So, often we have a shotgun approach.  Dr.

 

      DeLuca was sort of raising that question, why do we

 

      ask for three or four dissolution media sort of

 

      blindly?  If we had an understanding of what the

 

      physicochemical aspects were, if we knew what

 

      dissolution media or conditions would be most

 

      discriminating why would we ask for more?  We

 

      wouldn't.  We shared that.  And, the rule of thumb

 

      that I shared with you, which is a 30 year old rule

 

      which we still do not practice in industry is when

 

      you have certain acids or bases, however sensitive

 

      PK is, 4-6 or 3-6, go with the PK8 value.  That is

 

      the most discriminating.  That is a well

 

      established rule of thumb but we don't utilize

 

      that.

 

                So, often it is the shotgun approach, 3-5

 

      different media.  We just focus on pH.  Now, the

 

      surface tension of the dissolution media is about

                                                               224

 

      70/cm.  The surface tension of the GI fluid is

 

      about 30-50.  It makes a big difference.  So, why

 

      aren't we doing it at that surface tension?  So, it

 

      opens up all the questions that Lawrence raised.

 

                In practice, I think the frequent tendency

 

      is to utilize 0.1 normal HCL and I illustrated the

 

      concern from the perspective of the Japanese.  If

 

      that is a real concern, then we have to think about

 

      how to address that.

 

                Quality assurance versus in vivo relevance

 

      debate, I think that will be part of the decision

 

      tree process.  But one aspect that we have to think

 

      about is mechanistic understanding.  We haven't

 

      defined it.  And, I am proposing that we will use

 

      the ICH Q6A concept that we have already accepted,

 

      and here is that concept.  For example, this is

 

      section 3.3.2.3 of ICH Q6A, particle size

 

      distribution testing may also be proposed in place

 

      of dissolution testing when development studies

 

      demonstrate that particle size is the primary

 

      factor influencing dissolution.  So, if you are

 

      able to make that decision for a parenteral

                                                               225

 

      preparation why don't we make that decision for an

 

      oral preparation?  the reason here is because the

 

      parenteral suspension is a suspension.  In a solid

 

      dosage form you have compaction, and so forth, but

 

      we have technology that can even address that so

 

      this becomes a meaningful way to move forward.

 

      But, again, as was pointed out, the particle size

 

      distribution should be well characterized and well

 

      represented.

 

                So, mechanistic understanding as a

 

      proposal is that identification and scientific

 

      justification of causal, physical or chemical

 

      relationship between pharmaceutical materials

 

      and/or process factors that impact quality.  Here I

 

      want to draw a distinction.  Establishment of

 

      correlation may not be causal.  So, we want to be

 

      careful and that is the reason why in the PAT

 

      guidance we said correlation may not be sufficient.

 

      We need causality and that is process

 

      understanding.

 

                I think this is a significant debate and I

 

      think we may be able to achieve it in the U.S. but

                                                               226

 

      it has to occur in this.  Specifications equals

 

      standard.  That is the philosophy now.  That is the

 

      reason why release tests are USP market standard.

 

      Now, this is well ingrained.  I would propose we

 

      need to start thinking about changing that.  Market

 

      standards have a value, as Tom pointed out.  Market

 

      standards are fine.  You need them from the

 

      perspective that Tom laid out.  But then the

 

      release specifications and control studies should

 

      be different and be risk-based.  And this is the

 

      challenge.

 

                So, specifications are standard.

 

      Non-conformance means you have to reject or recall

 

      a lot.  Now think about this, most companies have

 

      this but we haven't utilized it.  You have to start

 

      thinking about a control limit with a target value

 

      and an acceptance range around a common cause of

 

      variability, with special causes of investigation,

 

      special causes to investigate.  So, if you think

 

      about this, one of the reasons why the concern is

 

      if you start reducing your variability the

 

      regulators around the world will say you have

                                                               227

 

      narrowed your specification is this phenomenon

 

      because, if you don't, they may not investigate and

 

      if there was a special cause that might be a signal

 

      for something else.  So, if you move towards a

 

      control philosophy and if you bridge this gap

 

      between CMC and GMP this is a way forward.  But,

 

      again, there is no consensus around the world on

 

      that.

 

                Step six, general consideration for

 

      identifying and developing statistical procedure.

 

      In my presentation I also identified the recent

 

      contribution from the USP.  I think it is a step in

 

      the right direction.  But it is, again, very

 

      similar to the parametric tolerance interval test

 

      concept and we have been debating that for at least

 

      the three years that I have been involved; I think

 

      it is more than that.  And, you will hear a

 

      progress report on that tomorrow from Bob O'Neill.

 

                This is where I really need the

 

      committee's help.  We have to start thinking

 

      differently.  Testing a hypothesis on every

 

      production batch is not the right way of thinking

                                                               228

 

      about this when you are in a state of control.  You

 

      are not testing every production batch as a

 

      hypothesis.  The gap between CMC and GMP I think is

 

      validation.  Hypothesis testing--my proposal would

 

      be to be limited to the validation part of it, and

 

      you really test the hypothesis that you have

 

      transferred the technology appropriately, and so

 

      forth.  So, specification setting and standards and

 

      hypothesis testing, such as parametric or

 

      non-parametric tolerance interval--and we have been

 

      working on that.  This is a recent hypothesis

 

      structure.  There was a recent proposal from our

 

      Office of Biostatistics for dissolution.  It should

 

      be limited as the time of approval and validation

 

      that our CMC reviewers make that call, and so

 

      forth.

 

                But then in production you have to move

 

      towards control.  So, you have to start thinking

 

      about control charts of variable, not attributes,

 

      where you really have a focus and target value and

 

      risk-based upper and lower limits; process

 

      capability analysis, not hypothesis testing on

                                                               229

 

      every lot.  This is important.  This is a very

 

      important principle on which I need your help.

 

                So, I will stop here.  The questions are:

 

      Are the steps outlined consistent?  Any additional

 

      steps you might recommend?  How should we prepare

 

      to come back to you?  What other scientific

 

      evidence is necessary to support this?  General

 

      consideration for identifying and developing

 

      statistical procedures and any other thoughts or

 

      comments you have on improvements.

 

                Committee Discussion and Recommendations

 

                DR. COONEY:  Ajaz, thank you.  I would

 

      like to take some time now for the committee to

 

      raise questions and discussion.  We will probably

 

      break at some point, have a chance to think things

 

      over and then come back and have more time for

 

      discussion, especially given the importance of the

 

      recommendations that you seek us to make.  Nozer,

 

      you had your hand on that button quickly.

 

                DR. SINGPURWALLA:  I have had it ever

 

      since he stood up.

 

                [Laughter]

 

                DR. COONEY:  The floor is yours.

 

                DR. SINGPURWALLA:  Well, I think you have

 

      come a long ways from the last meeting when I

                                                               230

 

      believe there was a discussion on mechanistic

 

      considerations.  What I would like to add is,

 

      Lawrence, you put up a differential equation.

 

      Remember that?  You put up a slide; you put up a

 

      differential equation.

 

                DR. YU:  Differential equation, right.

 

                DR. SINGPURWALLA:  Now, what struck me

 

      when you put it up is certain things.  The first

 

      thing is you didn't define everything, which is a

 

      bad way to put up an equation but I won't punish

 

      you for that.

 

                [Laughter]

 

                But what Ajaz said has some relevance to

 

      what you said.  That differential equation you put

 

      up is a mechanistic equation.

 

                DR. HUSSAIN:  Yes.

 

                DR. SINGPURWALLA:  It is a mechanistic

 

      phenomenon.

 

                DR. HUSSAIN:  Yes.

 

                DR. SINGPURWALLA:  And there is

 

      variability.

 

                DR. HUSSAIN:  Yes.

 

                DR. SINGPURWALLA:  And one of the elements

 

      of your equation was the particle size.  We have

 

      had some discussions here about the particle size

                                                               231

 

      not being a variable entity.  Therefore, one of the

 

      things I would recommend doing is--you asked what

 

      additional scientific evidence you need--I would

 

      encourage you to look at that particular equation

 

      and make it stochastic so that it becomes a

 

      stochastic differential equation, and I would use

 

      whatever knowledge you have about particle size

 

      distributions, and my colleague from Pfizer has

 

      given me some clues about what could be particle

 

      size distribution, and somebody else also.  I would

 

      like to suggest that you merge the two, the

 

      deterministic, mechanistic equation and the

 

      particle size equation.

 

                The comments you made about correlation

 

      and causation are germane and correct.  But the

 

      reason is this, that correlation only measures

                                                               232

 

      linear relationships and is not indexed by time.

 

      Causation is a time index phenomenon.  If I smoke I

 

      will get lung cancer, assuming that that is the

 

      causal.  So, there is a time phenomenon.

 

                One of the slides that you put up and you

 

      quickly slid by is wrong.  That hypothesis.

 

                DR. HUSSAIN:  As an example--

 

                DR. SINGPURWALLA:  That is wrong.

 

      Somebody has to fix it.  I wouldn't put it up

 

      again.

 

                DR. HUSSAIN:  You have to tell that to Bob

 

      O'Neill tomorrow.

 

                DR. SINGPURWALLA:  No way!

 

                [Laughter]

 

                DR. HUSSAIN:  Thank God, it was not mine.

 

                DR. COONEY:  But, Ajaz, you have been

 

      adequately warned!

 

                DR. HUSSAIN:  Yes.

 

                DR. SINGPURWALLA:  I will tell you what

 

      the problem with that equation is.  You have two

 

      probabilities there.  You have the null hypothesis.

 

      You are making hypotheses on probabilities of

                                                               233

 

      certain events.

 

                DR. HUSSAIN:  But that is the basis of the

 

      parametric tolerance interval test that you will

 

      hear tomorrow.

 

                DR. SINGPURWALLA:  Maybe I should stay at

 

      home!

 

                DR. COONEY:  Ken?

 

                DR. MORRIS:  I mean, I think this is

 

      clearly the right track and perhaps overdue,

 

      hypothesis testing aside for the moment, but the

 

      question I have is, is it within the scope of what

 

      we will all jointly do to say that the dissolution

 

      test, for example, is development activity as

 

      opposed to a manufacturing control for either bio.

 

      or control of process development?

 

                The other thing is that if the decision

 

      tree--I can't remember which one it was now--where

 

      you had mentioned the design space section for

 

      development, that is really the whole enchilada

 

      here.  Right?  I mean, in a sense, if we do that we

 

      have done everything.

 

                DR. HUSSAIN:  Right.

 

                DR. MORRIS:  With respect to laying out

 

      quality by design development.

 

                DR. HUSSAIN:  Yes.  No, I think, Ken, in

                                                               234

 

      many ways the decision trees should sort of focus

 

      on not directing what to do but more in terms of

 

      what are the key questions that need to be asked

 

      and direct what the decision should be.  So, that

 

      is the only way we can avoid interfering with the

 

      development program.  We don't want to do that.

 

      So.

 

                DR. COONEY:  The use of the decision trees

 

      acknowledges the fact that not all drug products

 

      behave in exactly the same way--

 

                DR. HUSSAIN:  Yes.

 

                DR. COONEY:  --in a standard system, and

 

      not all drug products act physiologically by the

 

      same mechanism.

 

                DR. HUSSAIN:  Right.

 

                DR. COONEY:  As I understand what you put

 

      forward, these decision trees would allow you to

 

      have a standard process to develop methodologies,

 

      but not constrained to always having a product work

                                                               235

 

      the same way.

 

                DR. HUSSAIN:  Right.

 

                DR. DELUCA:  Your number three there--I

 

      think you developed the first two very nicely.

 

      Number three, what additional scientific

 

      evidence--you know, once the plan is outlined, I

 

      think that is the decision tree.  Once the plan is

 

      outlined and the steps are rationalized then, you

 

      know, the additional evidence that is needed will

 

      surface.

 

                DR. HUSSAIN:  Yes.

 

                DR. DELUCA:  And I think that will surface

 

      from the decision tree.

 

                DR. HUSSAIN:  Yes.

 

                DR. DELUCA:  One thing I want to say about

 

      knocking out the dissolution test--

 

                DR. HUSSAIN:  We are not; we are not.

 

                DR. DELUCA:  No, I know but I am saying,

 

      you know, when you talk about particle size

 

      distribution in parenterals for a suspension that

 

      is very straightforward.

 

                DR. HUSSAIN:  Yes.

 

                DR. DELUCA:  I mean dissolution is going

 

      to be related to particle size.

 

                DR. HUSSAIN:  Yes.

                                                               236

 

                DR. DELUCA:  But you are talking about an

 

      order of magnitude of particle size distribution

 

      here a lot different.

 

                DR. HUSSAIN:  Sure.

 

                DR. DELUCA:  I mean, you are in an oral

 

      form and, in fact, I am not even sure that in the

 

      parenteral size distribution is as important as

 

      having a minimum particle size.  You have to be

 

      below a certain particle size; you have to get it

 

      through the needle.  So, the thing is it has to be

 

      very small and so that makes a difference and it is

 

      related then, so you don't need to do a dissolution

 

      test if you have a low enough particle size.  But

 

      with the oral products you have so many other

 

      factors involved--

 

                DR. HUSSAIN:  No, that is the reason I

 

      sort of clarified that I used that as an example to

 

      construct my statement of what mechanistic would

 

      be.  But the key aspect was, in a sense, if you are

                                                               237

 

      able to achieve a state of control with some of the

 

      new technologies--in fact, Jerry, at a science

 

      forum a couple of days back, presented an imaging

 

      approach to looking at dissolution prediction.  I

 

      didn't mention that to him but it completely

 

      coincides with the percolation theory that Hans has

 

      been progressing and just by looking at that you

 

      could have said that.  The purpose of that was to

 

      simply construct a statement of mechanistic basis

 

      for a decision to move forward.  So.

 

                DR. COONEY:  Tom?

 

                DR. LAYLOFF:  I think it is quite striking

 

      that we are having this discussion now.  You know,

 

      it has been about 30 years since we started looking

 

      closely at content uniformity and putting market

 

      standards on that, and looking at dissolution as a

 

      means to assure bioavailability.  It has been 30

 

      years.  And, I think that one of the things that we

 

      have seen is what Cindy was saying, FDA likes

 

      method 1 and 2; FDA likes content uniformity

 

      testing, likes dissolution testing.  So, the

 

      industry, instead of focusing on quality systems,

                                                               238

 

      directed research and formulas and production,

 

      actually kept that focus on a 30 year-old concept

 

      that dealt with the market issues 30 years ago

 

      instead of building the quality systems that deal

 

      with the present.  I am happy to see those other

 

      ones go away and let's go on with quality systems.

 

                DR. COONEY:  Art?

 

                DR. KIBBE:  Yes, since we are really deep

 

      into discussing particle size, let me throw out

 

      that really the key ingredient in understanding

 

      dissolution is surface area measurements and not

 

      particle size, and that particle size distribution

 

      means different things to different kinds of

 

      formulators at different times and we have a really

 

      good understanding of the kinds of particles we

 

      want when we compress a tablet and what percent of

 

      them are fine, and so on.  So, it is a different

 

      game.

 

                One of the issues that comes into play is

 

      not only does the particle size matter but if you

 

      have too small a particle size you start to get

 

      thermodynamic forces acting to cause aggregates,

                                                               239

 

      which give you a larger particle than you started

 

      out when you measured it the first time.  So, what

 

      I am getting to is that the issue is complex and

 

      the controls have to be put on for each system in

 

      each situation, taking into account what we know

 

      theoretically, and not impose as a general rule

 

      that just sits on top of everything.

 

                DR. COONEY:  Ajaz, one of the

 

      implications, as I understand it, of the proposal

 

      is that one would develop methods based around

 

      understanding of the science, the mechanisms that

 

      are controlling the important phenomena.  This

 

      suggests that the responsibility goes right back to

 

      the developer--the developer, CMC and the

 

      manufacturer, cGMP, and you are going to bring them

 

      together.  But this responsibility goes back to

 

      them to identify what those mechanisms are as

 

      opposed to simply adhering to a standardized assay

 

      of some type.  This is the implication as I see it,

 

      which will change the way the development work is

 

      done in the first place.

 

                DR. HUSSAIN:  It may change.  I think

                                                               240

 

      maybe the industry colleagues--

 

                DR. COONEY:  That was an invitation, Gerry

 

      and Paul, to respond.

 

                MR. MIGLIACCIO:  I mean, it is consistent

 

      with the ONDC restructuring.  It is consistent with

 

      getting away from the check-box mentality of the

 

      CMC section to put the appropriate science into the

 

      CMC section.  So, yes, it is going to change the

 

      way you do things and, hopefully, it will change

 

      the overall process of reviewing, approving and

 

      then continuous improvement around our products.

 

      So, yes, there is no doubt about it.

 

                I guess, since I have the microphone, why

 

      tactical step two?  What is behind your question

 

      around currently marketed products?  Because while

 

      quality by design for new products is starting to

 

      be a reasonably well understood concept, you cannot

 

      go back redesign a marketed product by those

 

      principles.  There is a lot we can do with

 

      currently marketed products so why did you say

 

      currently marketed products, question mark, when it

 

      comes to tactical step two?

 

                DR. HUSSAIN:  So, you picked it up!  Well,

 

      I think clearly there is a hesitation there, and

 

      the hesitation comes from the fact that what we

                                                               241

 

      have here--the challenges we have are destructive

 

      test and the hesitancy is variability of units in a

 

      pivotal clinical batch to be used is a key concern.

 

      So this is on the development side right now.  And,

 

      in a sense, what you do is you declare this as

 

      acceptable.  So, in the current scenario our

 

      specifications might be tighter than the capability

 

      of that and that is the means for imposing

 

      continuous improvement.

 

                So, the development information and the

 

      stability of the batch really overcomes this

 

      hesitation.  That essentially is the key here.  For

 

      step number two, one of the conditions that becomes

 

      is for this approach the clinical pivotal lot or

 

      the bio. lot must be stable--stable, I am talking

 

      about consistent--and its variation understood to

 

      the extent that the unit may be sampled for a

 

      destructive gauge R&R.  That is the fundamental

 

      principle of a destructive gauge R&R.

 

                Now, we don't have this information for

 

      currently marketed products so that was the reason

 

      for the question mark.  Can you overcome the

 

      hesitancy without that?  But I did put that, and so

 

      forth, to illustrate that I think we need to move

 

      in that and do it for those also.

                                                               242

 

                Here is an example of what I mean by

 

      stable process.  Is this a stable process?  This is

 

      actually real data from a company.  A friend of

 

      mine sneaked it to me.  So.  The percent

 

      dissolution as a function of manufacturing time,

 

      and you see that in this case this was

 

      non-homogeneous distribution of an excipient, which

 

      we never check, was the cause of this.  But most of

 

      these are never caught.  We approve those; they are

 

      part of the system.  I can show you many examples.

 

      Validation may not always catch this because the

 

      variation, as Gerry you pointed out, you learn more

 

      in manufacturing than anything else.  So, for the

 

      gauge R&R step two to occur, you will need to

 

      demonstrate complete stability of this lot.

 

                DR. COONEY:  Paul?

 

                DR. FACKLER:  To think about the

 

      implications of what is being discussed here for

 

      generic products in particular, it is gratifying to

 

      hear that FDA recognizes that dissolution

 

      parameters really are based on a formulation and

 

      very often, in fact most of the time, generic

 

      products have a different set of excipients than

 

      the brand product that they are bioequivalent to.

 

      It makes one wonder then why the generic company

                                                               243

 

      would be held to the same dissolution

 

      specifications that the brand product, with a

 

      different set of excipients and different

 

      formulation, might be held to.

 

                So, I support a revisit of the whole

 

      dissolution concept and, frankly, it is something

 

      we have struggled with.  We have a bioequivalent

 

      product, for instance, yet can't meet dissolution

 

      specs based on a different set of excipients.

 

                DR. HUSSAIN:  No, I did allude to that

 

      fact in the background paper that you might have

 

      seen.  Dissolution specification is such a complex

 

      performance test that depends on many factors, such

                                                               244

 

      as hydrodynamics.  If it appropriate for one

 

      formulation design, it may not be appropriate for

 

      the second.  That creates that.  In absence of

 

      additional information such as further development

 

      to understand how the formulation might behave, we

 

      have very limited choice--I mean, that is a concern

 

      that you see from the regulatory side.  The way we

 

      approaching it I think we can alleviate that

 

      concern and actually address that scenario, and

 

      this is one of the reasons to address that scenario

 

      too.  So, you are right.

 

                DR. COONEY:  Marvin?

 

                DR. MEYER:  Ajaz, not being familiar with

 

      this area at all, let me ask a question anyway.

 

      How easy is it going to be in all cases or all

 

      complex formulations to develop a total mechanistic

 

      understanding of the behavior of a given product,

 

      or will we sometimes have to say, well, we made a

 

      decent effort; let's go back to the old dissolution

 

      and whatever?

 

                DR. HUSSAIN:  No, I think you will not

 

      have a full level of mechanistic understanding, and

                                                               245

 

      so forth, but if you approach it right from the

 

      beginning and you start building it so you

 

      development information really will add to that.

 

      So, from a control strategy, in most cases you will

 

      not have complete mechanistic understanding but

 

      then at least you have enough understanding to put

 

      in the right controls, and so forth, and not rely

 

      on one particular test at the end.  So, I think it

 

      becomes a combination of the level of complexity

 

      you have and the degree of uncertainty you have

 

      that provides a means to say this is the control

 

      strategy.  So, you have to marry or arrange your

 

      control strategy in light of the complexity and in

 

      light of the uncertainty that is present with

 

      respect to the intended use of the product.  So.

 

                DR. MEYER:  I can certainly see from a

 

      manufacturing point of view that a total

 

      understanding of the product and the process would

 

      be great, but it is also great to have a product on

 

      the market that works.

 

                DR. HUSSAIN:  Sure, exactly.

 

                DR. MEYER:  So, the agency isn't proposing

                                                               246

 

      that ultimately down the line I am sorry, you

 

      didn't identify--

 

                DR. HUSSAIN:  No, no at all.

 

                DR. COONEY:  What I would like to suggest

 

      at this point is that we take a break for 20

 

      minutes--this is by design, an extra five minutes

 

      but this comes with a price.  As we think about the

 

      questions that have been put before us, the six

 

      questions, I would like for the committee to think

 

      about, as they are sipping their coffee,

 

      particularly questions two and three, what

 

      additional steps or changes would you recommend to

 

      improve what has been suggested?  And, three, what

 

      additional scientific evidence is necessary to

 

      support the development and implementation of the

 

      plan?

 

                The reason for the extra five minutes is

 

      because I think it is important that we think about

 

      the impact of the recommendations that we are going

 

      to address later today, beyond the discussion, are

 

      there unintended consequences?  What are the

 

      impacts that we need to think about, not that we

                                                               247

 

      need to identify them all, but what are the things

 

      we need to think about as we go forward?  So, for

 

      that an extra five minutes of coffee break.  Let's

 

      reconvene at about 3:12.

 

                [Brief recess]

 

                DR. COONEY:  I would like to welcome

 

      everyone back.  We have a period now where we can

 

      have a discussion around the questions that are on

 

      the table.  The questions that we have been asked

 

      to consider are summarized on the screen.  What I

 

      would like to do in the next hour approximately is

 

      to have an open discussion amongst the committee on

 

      these questions.  I would like to get to a point

 

      where we can take a vote as a committee in terms of

 

      voting either--I will ask the voting members of the

 

      committee to support the recommendations of the

 

      committee going forward.  I will do that at the

 

      end.  It will be yes, no or abstain.  You will be

 

      given options.  The voting members of the committee

 

      will be the committee plus the consultants that are

 

      here.

 

                But prior to that, I think the request

                                                               248

 

      that we have on the table is a plan that is laid

 

      out on the slide that is before us that has a

 

      series of steps.  The first, and I think the

 

      central question, is are the tactical steps

 

      outlined--and these are outlined in slides 8

 

      through 15 of Ajaz's presentation--are they

 

      consistent with the goal of quality by design?  I

 

      thought we might begin by talking about these

 

      tactical steps first, have some discussion around

 

      that, and then to work through the other questions

 

      that we have been given.  It is particularly

 

      appropriate and important for the committee members

 

      to take this opportunity and share with Ajaz and

 

      the rest of the team their ideas and thoughts, and

 

      it is my understanding that, if these

 

      recommendations are approved, what it represents is

 

      a step forward; the questions that have been

 

      outlined will be examined and possibly as early as

 

      our next meeting specific recommendations for our

 

      discussion will be brought forward.

 

                With that, let's take a look at question

 

      one which really revolves around the tactical

                                                               249

 

      steps, slides 8-15, that we have in front of us.

 

      Then perhaps we can go through these steps with an

 

      open discussion around them.  Ajaz, could you go to

 

      your slide 8, which is tactical step one?  This is

 

      really the first two steps in this plan.  Given the

 

      limitations of the dissolution assay as currently

 

      practiced, and its relationship or lack thereof to

 

      therapeutic efficacy and safety, to look at

 

      alternative suitable methods and strategies in

 

      order to evaluate the quality of drug products.

 

                DR. SINGPURWALLA:  Let me just get the

 

      ball rolling.  Steps one and two essentially

 

      encapsulate what you have in mind in the boxed

 

      items:  Information collected should facilitate a

 

      shift from the deterministic to a probabilistic

 

      design culture.  That is true for step one and step

 

      two.  And, I don't see any reason why we should not

 

      endorse it.  It is the natural thing to do and my

 

      particular position on this is to go ahead and

 

      endorse it, at least step one and step two, as I

 

      see it.

 

                DR. COONEY:  Let me also put on the table

                                                               250

 

      the question are there suggestions, modifications

 

      that one might make to these steps?  We can talk

 

      about that as we go through this as well.  Gerry?

 

                MR. MIGLIACCIO:  Yes, I mean, the whole

 

      discussion today and steps one and two really focus

 

      on dissolution.  I guess I thought what we were

 

      trying to get to is a more scientific measure of

 

      performance, process performance being the quality

 

      assurance measurement and product performance being

 

      the in vivo.  So, do we need to start with that

 

      decision criteria first, Ajaz?  I mean, the

 

      tactical steps are all focused on dissolution

 

      without saying is it really dissolution that we

 

      should be talking about.

 

                DR. HUSSAIN:  Clearly, I think the

 

      thoughts we had--why is step one the first step,

 

      and I think that is the key.  The way we see it is

 

      the current dissolution test system, as we use it,

 

      is an essential decision tool during product

 

      development and for regulatory decisions so we have

 

      it right now.  So, step one and two are overcoming

 

      some of the challenges that we have.

 

                Gerry, what you are suggesting--we are not

 

      eliminating that target at all and what we are

 

      doing here is improving upon one tool that we are

                                                               251

 

      using currently, and we anticipate using currently

 

      and in the future also, improve it and then also

 

      work on other ones, and other decision trees that

 

      we are planning will build in aspects that I think

 

      you are alluding to.  So, our thought process of

 

      why is step one the first step is that we

 

      anticipate dissolution testing--that the

 

      methodology will be with us for a long period of

 

      time, and here the uncertainty with respect to the

 

      suitability criteria and how we set acceptance

 

      criteria--there seems to be a disconnect here and

 

      there is a lot of frustration around these issues.

 

                So, step one and two combined address the

 

      immediate need, and also set up a system which is

 

      more rugged, we can be more confident to start

 

      building alternate methodologies.

 

                MR. MIGLIACCIO:  So, we are really talking

 

      about more scientific approaches to dissolution.

 

                DR. HUSSAIN:  Step one and two, yes.

 

                MR. MIGLIACCIO:  I understand gauge R&R

 

      well but I am not an expert.  Can you get enough

 

      information out of a gauge R&R on a single bio.

 

      batch?

 

                DR. HUSSAIN:  One of the key aspects that

 

      we are trying to get from this is that in most

                                                               252

 

      simple cases I think we think the study will not be

 

      a full-blown gauge R&R, and so forth.  We think

 

      that product development information will guide you

 

      through that.  Now, there are elements and there

 

      are arguments out there that you need to know how

 

      sensitive your particular formulation is to the

 

      conditions of the dissolution test.  So, from that

 

      aspect, I think this will allow us to gauge the

 

      sensitivity of your formulation, your particular

 

      formulation to the chosen dissolution method.

 

      Hopefully, if it is done early you would remove

 

      that, and so forth.  This actually then becomes

 

      simply a study to benchmark variability.  So, that

 

      is the reason I anticipate that for most simple

 

      cases this may not be an extraordinary effort

 

      necessary to really do a full-blown gauge R&R.

 

                MR. MIGLIACCIO:  But you don't get the

 

      batch-to-batch variability.

 

                DR. HUSSAIN:  Correct, but at least you

 

      benchmark the product and then you could get

 

      additional benchmarks from your validation batches,

 

      and so on and so forth.

 

                DR. COONEY:  Art?

 

                DR. KIBBE:  I agree with my statistician

 

      friend that we probably ought to move forward, but

                                                               253

 

      I think Gerry raised a really important point, and

 

      I was going to raise it too, that is, this whole

 

      concept really started several years ago when we

 

      started really talking about what is the essential

 

      information we need to know to make sure that we

 

      have good quality products for the citizens.  That

 

      started with PAT and it kept going.  And, this

 

      really is just a natural evolution of the

 

      regulation of the quality of the process that gives

 

      us pharmaceuticals for sale in the United States or

 

      anywhere.

 

                So, as long as we, as a committee, and the

 

      FDA, with you as its spokesperson, understand that

                                                               254

 

      this is just one more small step moving forward and

 

      not a whole process--I mean, we are not about

 

      dissolution testing; we are about eliminating

 

      unnecessary testing and doing the correct testing

 

      to make sure we have quality products.  That is

 

      one.

 

                Second, the tremendous variability in

 

      human response to a given drug product is just like

 

      a thunder cloud on top of a small camp fire of

 

      dissolution testing.  And, if we get better and

 

      better at controlling the process and if then the

 

      batch is slightly out of that control, that doesn't

 

      justify not selling that batch because it is well

 

      within the goal posts that we have been working

 

      with from the beginning.  So, I think we need to

 

      also fold into here the understanding that as the

 

      process gets under tighter and tighter control

 

      deviations from that control must be investigated

 

      and must be understood but aren't necessarily a

 

      justification for not releasing the batch.  I don't

 

      know how you fold that in.  When we start talking

 

      about dissolution as a terminal test, whether we

                                                               255

 

      have surrogate markers that track the process and

 

      tightly control it, and if we are a little bit out

 

      we can allow the company to do a quick extra test

 

      and say, yes, it is still good enough but we are

 

      going to find out why it is out.  If we don't, we

 

      are going to scare off half the companies from

 

      following us down this path to really tight control

 

      systems.

 

                DR. HUSSAIN:  Your first point is well

 

      made and in the break I was asked to sort of

 

      summarize that, and you just did that for me.  But

 

      the second point is more challenging.  If your

 

      release test right now is a compendial test, that

 

      is a market standard.  You have no room for

 

      anything there.  That is the law.  So, that is the

 

      law.  So, how you sort of address that I think is a

 

      much larger issue.  In the PAT guidance, if you

 

      recall, we actually suggested in a sense that there

 

      has to be a way for moving forward and we created a

 

      system which we call research data so if it is an

 

      alternate procedure that you are using that is not

 

      your decision based on your compendial, you have a

                                                               256

 

      way forward under the PAT guidance for that right

 

      now.

 

                But the point is, in a sense, for

 

      standards you have to draw the line somewhere and

 

      compendial standards draw the line.  You have to

 

      conform to that.  That is the law.  So, the whole

 

      strategy then is that I think you have to move

 

      towards what I would like to sort of share with

 

      you, a concern that you expressed, and it is an

 

      important concern--if I can find my slide--that I

 

      think goes to another dimension.

 

                I think we are moving towards a state of

 

      control and demonstrating a state of control is

 

      important but, at the same time, for some processes

 

      we will never reach the state of assured

 

      statistical control.  So, we have to bring a

 

      risk-based decision to that in the sense of how do

 

      you sort of manage that because every deviation is

 

      investigated.  So, there is an element that we do

 

      intend to discuss, and discuss internally as well

 

      as I think in workshops, and so forth, the need to

 

      debate engineering control versus process control. 

                                                               257

 

      There is an aspect to that.

 

                DR. COONEY:  Art, let me see if I have

 

      heard your point clearly, that is, you are arguing

 

      or you are suggesting that it is very important to

 

      think through how this additional information, how

 

      this different information will, in fact, be used

 

      from a regulatory perspective.

 

                DR. KIBBE:  Yes, I think we have to be

 

      careful, because we can narrow the goal posts, that

 

      we don't necessarily do it if it doesn't gain us

 

      anything clinically.  We have talked about this

 

      over and over again.  The first point I made is

 

      that this is just one more step in a process that

 

      started several years ago and I think it is long

 

      overdue, but we shouldn't focus only on dissolution

 

      as the only thing we are doing because we are doing

 

      lots of other things too.

 

                The second is that in order to get the

 

      companies to come along with us, they can't view it

 

      as an opportunity for the regulatory agency to

 

      nit-pick them.  We talked about that before and I

 

      just wanted to make sure it was kind of restated

                                                               258

 

      for the record.  That is all.

 

                DR. COONEY:  That is an important point

 

      and that is why I wanted to get clarity, and it

 

      should be part of the recommendation going forward.

 

      Ken?

 

                DR. MORRIS:  Yes, to your scient point,

 

      Art, and I think this may be a little bit out of

 

      sequence, in my sort of image the way things would

 

      ultimately be dissolution in some form--although I

 

      would argue that there may be better forms and that

 

      is a different discussion for the next question, I

 

      guess--that is a develop tool.  These are part of

 

      the critical quality attributes that you want to be

 

      able to build in with enough significance to have

 

      faith in it.  But then you are really in the

 

      position of formulating the process variables that

 

      are dictated by, hopefully, the first principles to

 

      determine your design space.  Once you have the

 

      design space, then you are taking advantage of the

 

      PAT guidance and others to do real-time release,

 

      hopefully, but at least you will be releasing

 

      within a parameter space that says if I control to

                                                               259

 

      my endpoint, which should be a manifestation of the

 

      proper attributes, then anywhere in that design

 

      space that it takes me to get to my endpoint is

 

      fine, and that is release.  You release the product

 

      based on the attainment of the endpoint as opposed

 

      to a release spec after the fact.  The only

 

      criteria are that you are, first of all, within

 

      your design space and that the design space has

 

      been developed so that it does accurately represent

 

      the process.

 

                DR. COONEY:  Tom and then Paul and Gerry?

 

                DR. LAYLOFF:  Yes, I don't think that we

 

      will see a change in the market standards, the

 

      limits that are there, because they are very

 

      appropriate for among lab assessments within the

 

      statistical bounds that you want to have for a

 

      release.  But the company release specifications

 

      would be something more like a control chart I

 

      think where you have the desired in the center and

 

      then the bracketing on the outside.  If you start

 

      drifting away from that you start looking at kappa,

 

      corrective actions to move it back down.  But if it

                                                               260

 

      goes outside those bounds, then you are probably

 

      going to miss the market standards and if you miss

 

      the market standards you have marketed an illegal

 

      product.  But I don't think the market standards

 

      are going to change.  If you look the among

 

      laboratory test criteria and the expected

 

      statistics, it is a very rational thing to do, to

 

      have limits like that.

 

                DR. COONEY:  Paul?

 

                DR. FACKLER:  I agree.  Of course, I

 

      endorse pursuing examining whether there is a

 

      better way to deal with dissolution.  But I still

 

      think fundamentally we need to ask why

 

      dissolution--is it to hold variability on the

 

      manufacturing process or is it to assure the

 

      patient that when he takes the product it is going

 

      to perform the way it is labeled to?  We do content

 

      uniformity for exactly that.  If the tablet is 10

 

      mg and you find that there are 7-8 mg you wouldn't

 

      want to release that lot.

 

                On the other hand, you do a dissolution

 

      test and you are left scratching your head, saying,

                                                               261

 

      well, I know that this dissolution test has nothing

 

      to do with the way it is going to dissolve and be

 

      absorbed by the particular patient, yet you are

 

      forced to scrap the lot.  So, really I would

 

      encourage you to keep in mind what the goal of the

 

      release test is above everything else.

 

                DR. HUSSAIN:  Point well taken, and please

 

      keep in mind that the decision trees that we are

 

      developing will actually address that aspect.  The

 

      key is this, we don't want to go with

 

      one-size-fits-all, like you need a dissolution test

 

      for everything.  That is not the intention.  The

 

      decision trees will sort of guide us through when

 

      it may be needed; when it may be not needed; and if

 

      it is needed for quality assurance how do you

 

      approach that; and if it is needed for, say,

 

      characterization of a product, say, post-change

 

      like we do in SUPAC, how would we do that?  If you

 

      are using it for biowaiver, as Lawrence suggested,

 

      can we think about more biorelevant conditions that

 

      might expand biowaiver decisions for BCS?  So,

 

      those would be the considerations.

 

                So, step one and two are not intended for

 

      that particular purpose but it sets up instrument

 

      suitability criteria that makes the system more

                                                               262

 

      stable for us to use, and it also provides an

 

      experimental approach to start think of

 

      characterizing the variability and doing more than

 

      just six tablets but characterizing the variability

 

      to start using that in our decision-making.  So,

 

      step one and two are just for that and all the

 

      other aspects that you sort of mentioned are for

 

      decision trees where we want to capture that.  So,

 

      that is how we want to approach it.

 

                DR. COONEY:  Gerry?

 

                MR. MIGLIACCIO:  Yes, I want to go back to

 

      Ken's comments.  Ken is always a visionary.

 

      Unfortunately, the vision is a ways out.

 

                DR. MORRIS:  Yes, six weeks.

 

                [Laughter]

 

                MR. MIGLIACCIO:  We might all be in

 

      rocking chairs by that time.  But, you know, he has

 

      raised a good point which I think an industry

 

      perspective needs to put out there, and that is,

                                                               263

 

      you know, coming in with greater understanding of

 

      product to process to come up with better

 

      performance measurements will work if, when we are

 

      sitting down and discussing these with the agency,

 

      it is not just that plus the layer of the

 

      traditional dissolution test and the traditional

 

      this and the traditional that.  If this is just

 

      layering on more and more, then no company will

 

      invest the resources to do it.  We will do it

 

      internally.  We will have our internal controls

 

      because that process understanding makes us more

 

      efficient, more effective, but this process will

 

      stop--Art has hit it right on the head, there has

 

      to be an understanding that by coming in with this

 

      we have to take a new path and not be mired in

 

      history.

 

                DR. MORRIS:  If I can comment just real

 

      quickly, I think the other thing is that, from the

 

      agency side, adding another layer isn't going to

 

      win any friends on the agency side either.  Right?

 

      You guys can speak for yourself but I think they

 

      are already stressed to the limit.

 

                DR. HUSSAIN:  No, I think we clearly

 

      understand that, and the key is, in a sense, if we

 

      keep holding on to things which may not be adding

                                                               264

 

      value and keep adding more the system doesn't work.

 

      So, the decision tree will have to capture a

 

      process which says these are the key questions that

 

      we need to address and this is how we will control

 

      this.  If your control strategy meets that, for a

 

      new product you might have a completely different

 

      set of specifications which are non-traditional.

 

      But keep in mind that if you already have approved

 

      products you have those locked in and if you have a

 

      compendial you have those locked in.  So, the

 

      future of specifications and controls could be very

 

      different.  So, we have to balance the two as we go

 

      along.

 

                DR. COONEY:  Nozer?

 

                DR. SINGPURWALLA:  Ajaz, I am going to

 

      comment on this slide.

 

                DR. HUSSAIN:  Please.

 

                DR. SINGPURWALLA:  There is no need to

 

      debate.  While this is a fictitious issue raised by

                                                               265

 

      some of my statistician colleagues about 10, 15

 

      years ago, what they did is if you have engineering

 

      control then there is no need to do statistical

 

      process control.  Engineering control essentially

 

      achieves control for you.

 

                DR. HUSSAIN:  Yes.

 

                DR. SINGPURWALLA:  This was some kind of

 

      an article written by some of my colleagues, not

 

      necessarily friends, in journals, trying to make a

 

      distinction, and the distinction is completely

 

      fictitious.  So, I don't think you should spend too

 

      much time debating this.  I think engineering

 

      control has elements of stochasticity in it and

 

      that is about it.  So, I would just not bother with

 

      this question any more.

 

                DR. HUSSAIN:  I appreciate that because I

 

      expect this to be a debate in the committee, and

 

      the way I think we have constructed the QA

 

      guidelines, especially how to define that, we

 

      actually have approached it from an engineering

 

      perspective.  But I do have to put it on the table.

 

                DR. COONEY:  Mike?

 

                DR. KORCZYNSKI:  As most of you know,

 

      pharmaceutical companies are very conservative in

 

      nature and as long as the dissolution testing

                                                               266

 

      appears in the USP, even though the USP may say one

 

      can use alternative methods, I think there is going

 

      to be a major reluctance to abandon dissolution

 

      testing.  It was already mentioned that a lot of

 

      the emergent technologies, in fact, are sort of

 

      relegated to segments of the process, not

 

      necessarily the total process.  So, somewhere I

 

      would think some type of guidance document could be

 

      available that cites opportunities relative to

 

      dissolution testing, and maybe begin to provide a

 

      bridge and have companies begin to think that, yes,

 

      they can abandon the method if they use these

 

      potential alternatives that are cited.

 

                DR. COONEY:  Gerry?

 

                MR. MIGLIACCIO:  Yes, speaking for the

 

      innovators, you know, where there is no USP

 

      monograph that we are dealing with, we obviously

 

      can come up with any performance method that is

 

      appropriate that I think many companies and FDA are

                                                               267

 

      evaluating.  I mean, the PAT guidance gives us the

 

      open door to evaluate these, and there are a number

 

      of them under evaluation now.  None of them have

 

      proven out but I don't think it is going to be that

 

      much longer before they are.  And then, I don't

 

      know that we need guidance.  I think the instrument

 

      manufacturers will sell it for us.  Once they

 

      understand that the methodology and the equipment

 

      that they are providing is a surrogate for

 

      dissolution, I think they will be out there

 

      marketing it very aggressively to the entire

 

      industry.

 

                DR. COONEY:  Mike?

 

                DR. KORCZYNSKI:  Well, I think large

 

      innovative companies all basically have the

 

      wherewithal to develop their own emerging

 

      methodologies and PAT systems, but there are many

 

      smaller companies that would look for guidance

 

      prior to, say, abandoning a USP methodology.  You

 

      know, we heard the words that, oh well, one can do

 

      an alternative test but there is the reality of the

 

      field inspector who may interpret that if you are

                                                               268

 

      USP labeled you should be following the USP test.

 

      That is why I think some companies we be real

 

      reluctant to abandon that testing.

 

                DR. HUSSAIN:  One aspect is, I mean, we

 

      are not asking for abandoning, and so forth.  We

 

      want to sort of move towards an appropriate control

 

      or measurement that is relevant for that.  I mean,

 

      that is the whole intention.  So, I think I have

 

      some hesitation to use the word abandon.  That is

 

      not the discussion.  I completely understand I

 

      think the concern of the field and I think this is

 

      one of the gaps which we are addressing as we are

 

      moving along.

 

                One of the bullets in Cindy's was training

 

      and I think one of her jobs after this is to put

 

      together a training program for our field

 

      inspectors, and so forth, and Mehul and others I

 

      think are really--I think the CMC and GMP aspect,

 

      especially in how the quality assessment system is

 

      involved in ONDC decisions.  ONDC decisions have to

 

      be shared with their colleagues and inspectors.

 

      So, I think we are putting a systems perspective to

                                                               269

 

      address all of these concerns so I think we do

 

      understand the concerns there.

 

                DR. COONEY:  I think the specific point

 

      being made here is one of being sure that there is

 

      adequate communication of the work product of this

 

      group to the broader community.  It doesn't

 

      necessarily need to be a guidance but certainly

 

      adequate communication so that it can be used.

 

                DR. HUSSAIN:  One of the reasons for

 

      putting a tactical plan--I mean, we could have

 

      taken our time and brought a proposal to the

 

      advisory committee but we felt that we wanted to

 

      bring the tactical plan to engage and actually have

 

      the committee debate and discuss this.  When we do

 

      bring a proposal, I think most of the committee

 

      will at least have been engaged in discussing this

 

      among themselves.

 

                DR. COONEY:  So far we have focused on the

 

      tactical plan steps one and two, although we really

 

      have moved into three through five, which are the

 

      decision trees, as well.  Without necessarily

 

      curtailing discussion on any of those, I would like

                                                               270

 

      to consider step six in your plan, which is general

 

      considerations for identifying and developing

 

      statistical procedures.  I wonder if any one on the

 

      committee might have any commentary on this point.

 

                DR. SINGPURWALLA:  Yes.

 

                DR. MEYER:  While he is formulating his

 

      Bayesian response could I just say your proposed

 

      steps, page two, and six bullets so I presume the

 

      last one was number six.

 

                DR. HUSSAIN:  No, those are sort of

 

      specific tactical, and the others are this meeting,

 

      ICH and so forth.

 

                DR. MEYER:  Anyway, with respect to page

 

      two, the sixth one, from a committee member

 

      perspective I think that would be an excellent way

 

      to present whatever you choose to change,

 

      side-by-side comparison of new and generic, and why

 

      the old wasn't as good as the new, and how you are

 

      not really layering one thing on top of another but

 

      you are replacing one thing with the other.  It

 

      would be extremely helpful I think to present the

 

      information that way.

 

                DR. HUSSAIN:  Yes.  Sorry, I think my

 

      slide had a different number.

 

                DR. SINGPURWALLA:  Getting back to step

                                                               271

 

      six--

 

                DR. HUSSAIN:  This one?

 

                DR. SINGPURWALLA:  Yes, on page eight.  It

 

      seems reasonable in the sense that in principle

 

      what has been outlined is very reasonable.  We may

 

      have to disagree on details and detail steps.  I

 

      think you want to use control charts for variables

 

      and not attributes.  That makes sense.  You lose

 

      information when you use attribute data.  Process

 

      capability analysis, yes.  You don't want to test

 

      every lot.  You don't want to focus attention on a

 

      piece by piece; you want to look at the broader

 

      picture.  Yes, you need tolerance intervals.  How

 

      you are going to get them is a different matter.

 

      And, maybe there are other things that you may want

 

      to throw in which don't come to my mind

 

      immediately, but I don't see why this is not a

 

      reasonable step so, again, I would endorse it.

 

                DR. HUSSAIN:  Well, I think the challenge

                                                               272

 

      is significant.  This is the fundamental aspect

 

      because tomorrow you have a proposal to actually do

 

      hypothesis testing on every batch, and we have been

 

      discussing that for the last three and a half

 

      years.

 

                DR. SINGPURWALLA:  What is the alternative

 

      to not testing every batch?

 

                DR. HUSSAIN:  No, I think as our CMC

 

      reviewers are finalizing the specifications, and so

 

      forth, and then if you need a hypothesis, that is

 

      where it occurs.  But then imposing a hypothesis

 

      test on every production batch and then deciding

 

      whether you met the hypothesis completely negates

 

      the systems of GMP philosophy which says this is

 

      validated and under control.  So.  But the

 

      parametric tolerance interval test is exactly doing

 

      that.  So, that is the reason I think this sort of

 

      comes back.  Gerry, do you see that point?

 

                DR. MORRIS:  I don't know if it matters,

 

      you were talking before, you know, that it is hard

 

      to meet Short's criteria but there are more

 

      advanced SPC or engineering control techniques than

                                                               273

 

      those I guess older ones.

 

                DR. HUSSAIN:  See, the control chart and

 

      the limits are connected to the hypothesis, and so

 

      forth.  They are not disconnected.  But then you

 

      are approaching it very definitely, the philosophy

 

      and the system that you have to put in place is a

 

      different system as opposed to hypothesis testing.

 

                DR. MORRIS:  That is what I am saying, if

 

      you used a more sophisticated control system or

 

      whatever you want to call it--a filter, doesn't

 

      that suffice?

 

                DR. HUSSAIN:  It should.  We can discuss

 

      that as we debate that.

 

                DR. SINGPURWALLA:  I am presuming that

 

      that is encompassed as general vocabulary.

 

                DR. HUSSAIN:  Yes.

 

                DR. SINGPURWALLA:  You know, even though

 

      it is called engineering, there is a lot of

 

      probabilistic thinking behind that.  So, I endorse

 

      those.  If these techniques are becoming

 

      old-fashioned, as my colleague says and he is

 

      right--

 

                DR. MORRIS:  Long-standing.

 

                DR. SINGPURWALLA:  Yes, long-standing, I

 

      think it is time to look at other things.

                                                               274

 

                DR. COONEY:  Are there any other comments

 

      on this first question which is on slide two, that

 

      is, the tactical steps which are outlined in slide

 

      three of Ajaz's presentation?  I think embodied in

 

      part of that are the questions on additional steps

 

      that you would recommend to improve the plan and

 

      additional scientific evidence.  I think we have

 

      already made a number of suggestions to that end.

 

                DR. HUSSAIN:  In particular, I just want

 

      to repeat what I think my understanding has been.

 

      One aspect was that I think we have focused on

 

      decision trees and really the science will evolve

 

      from decision trees.  In one aspect, one proposal

 

      was from Cindy, the calibration, mechanical

 

      calibration conditions that we are using in our lab

 

      is what we are using and that will be a

 

      recommendation to industry to use the same one.  Is

 

      that sufficient or does Cindy need additional

 

      information to make her case for that?  You could

                                                               275

 

      think about it that way also.

 

                DR. MORRIS:  I mean, I think my opinion is

 

      already pretty clear but I just don't see the

 

      calibrator tablets as being value added.  So, to

 

      that end, I would say the alternative then is to

 

      have a far more rigorous mechanical calibration, a

 

      real gauge R&R in the sense of the word as you

 

      proposed it.

 

                DR. HUSSAIN:  In addition, we are doing a

 

      gauge R&R study for a couple of products that we

 

      are evaluating.  So, I think we will bring an

 

      example of a gauge R&R, a real-life example.

 

                DR. BUHSE:  I think Ajaz was making the

 

      point--some of you asked what would you do with a

 

      gauge R&R?  You know, it is a destructive test.

 

      How would you carry it out?  And, I think that is

 

      what we are trying to figure out in our lab by

 

      doing a few of them and kind of seeing what we get

 

      and seeing how many tablets you actually have to

 

      run, etc., etc., to try to get a feel for how much

 

      work it would actually be to do something like

 

      this.

 

                DR. HUSSAIN:  Right, and since we are

 

      using a commercial product we hope to--because in a

 

      destructive test, when the sample is destroyed the

                                                               276

 

      selection of your reference material, which is your

 

      clinical lot, really has to be very carefully

 

      thought out and planned out and the conditions

 

      under which you judge that they are acceptable.

 

      That goes hand in hand with our approval decision

 

      anyway.  But we are using a commercial material to

 

      do this so I think it will raise some aspects and I

 

      think we will learn something about that.

 

                DR. COONEY:  Gerry?

 

                MR. MIGLIACCIO:  Well, since you ask the

 

      question on calibrated tablets, after the last

 

      industry study, which was in the background

 

      material, the one remaining issue is vibration.  Do

 

      you feel in the FDA labs that that is now well

 

      understood and controlled?

 

                DR. BUHSE:  We actually measure vibration

 

      and we feel that we have a control over it

 

      definitely, yes.  We can measure it without having

 

      to use a calibrator tablet.

 

                MR. MIGLIACCIO:  So, I think we all agree

 

      that if we had a reasonable measure of vibration

 

      and could control it that the calibrator tablets

 

      aren't necessary.

 

                DR. COONEY:  Art?

 

                DR. KIBBE:  Just a quick point, even if

                                                               277

 

      you didn't, the one that we are using as a tablet

 

      calibrator we would have to redesign that, and I

 

      think we are far better off with engineering

 

      parameters in the long run.

 

                One point, we never should feel married to

 

      any test.  I mean, many, many years ago the test

 

      for quality of digitalis whole leaf was the pigeon

 

      death test where we injected a pigeon with a

 

      macerated solution of whole leaf digitalis until it

 

      died and then we rated the number of units of

 

      digitalis efficacy on that.  We clearly don't do

 

      that today.  Perhaps the dissolution test as a

 

      quality control test ought to go with the pigeon

 

      death test.

 

                DR. COONEY:  Paul?

 

                DR. KIBBE:  See if you can follow that!

 

                DR. COONEY:  That is a tough one to

 

      follow!

 

                DR. FACKLER:  I just want to remind

 

      everyone of the difficult position the generic

 

      industry is in with USP requirements imposed.  You

 

      know, part of what we do is try to obtain FDA

 

      approval but we are also bound to USP

 

      specifications.  So, I would again encourage FDA to

 

      work as closely with USP as possible to get both

                                                               278

 

      organizations aligned such that we are not in a

 

      position where we can satisfy one and not satisfy

 

      the other.  The whole industry is really at a loss.

 

                DR. HUSSAIN:  Gary, do you want to

 

      respond?

 

                DR. BUCHLER:  Thanks, Ajaz.  We certainly

 

      can discuss with USP revising the requirements so

 

      they are in line with our new paradigm that we want

 

      to set up for quality.  Clearly, we do have a

 

      relationship with USP and I think we know a few

 

      people over there so we should be able to discuss

 

      things with them.

 

                DR. LAYLOFF:  A couple of things, first of

                                                               279

 

      all, the industry can submit methods to the USP and

 

      submit changes to the USP.  That is one option.  I

 

      don't think it should be FDA.  If FDA is very

 

      concerned that a method is not suitable to protect

 

      the consumer, the public health, then the FDA could

 

      issue a standard and override the USP.  They have

 

      done that in the past.  So, if the FDA has grave

 

      concern about protecting public health they can go

 

      over, but the industry can submit changes.  You

 

      said you have a product that fails the dissolution

 

      but is bioequivalent to the innovator.  It means

 

      that the dissolution test is not a good surrogate

 

      and you should submit an alternate one.

 

                DR. FACKLER:  And we do that.  The problem

 

      is the timing involved in getting a USP monograph

 

      changed, and waiting for that often puts us at

 

      tremendous disadvantage economically.

 

                DR. LAYLOFF:  You can also get from the

 

      USP a letter waiving that requirement pending an

 

      option.

 

                DR. MORRIS:  Can I ask what is the impact

 

      if calibrator tablets were deemed to be unnecessary

                                                               280

 

      by FDA?  You would still have to use them?  Is that

 

      what you are saying?

 

                DR. HUSSAIN:  Well, I think that is a GMP

 

      issue and I think we have other options of a

 

      compliance policy guide.  So, there are a number of

 

      mechanisms to think about that.  I think the words

 

      we chose were very carefully chosen, an alternate

 

      method.  In my description of the vector for

 

      desired state anything we do should conform to the

 

      current standards in a sense.  I think we are

 

      trying to achieve a level of quality so that when

 

      tested with minimal standards there should not be

 

      an issue.  So, that is the basic premise on which

 

      we have developed the tactical plan.  It says an

 

      alternate procedure.  So, that is an option too.

 

                DR. COONEY:  If there are no further

 

      questions or comments from the committee, what I

 

      would like to do is to consider moving forward with

 

      a recommendation from the committee to the FDA that

 

      the proposed regulatory tactical plan, as described

 

      in slide two, be adopted and moved forward,

 

      recognizing that our responsibility is to make a

                                                               281

 

      recommendation to the FDA, it will then be

 

      considered by the  FDA as to how it actually is

 

      implemented.

 

                I would also suggest that, in addition to

 

      the tactical plan, as described in slide two--

 

                DR. HUSSAIN:  This one?  Right?

 

                DR. COONEY:  No, that plan and then, in

 

      addition, the proposed steps in slide three are

 

      incorporated in this recommendation.

 

                I have heard from this discussion six

 

      other additional points and I will just read these

 

      quickly, acknowledging that this is one step in

 

      quality by design, not just a focus on dissolution

 

      testing.  That one needs to think through how the

 

      information will be used in both manufacturing and

 

      regulation.  That it is important to keep in mind

 

      the ultimate goal of the release test, which is the

 

      patient.  The implementation plan needs to consider

 

      the impact on the manufacturer and the regulator.

 

      That may be redundant with the point I made above.

 

      To develop a suitable communication strategy for

 

      the work to be done, and to also work through the

                                                               282

 

      implications to the generic products with respect

 

      to USP requirements.  These are additional factors

 

      that I have heard out of the conversation this

 

      afternoon.

 

                DR. HUSSAIN:  I think the next page, page

 

      four, had one more step which Prof. Singpurwalla

 

      really commented on.  So, we will include that in

 

      your recommendation.  Developing detailed procedure

 

      and harmonization--these are part of our tactical

 

      plan but not necessary for discussion at this

 

      stage.  So, if you would include this as part of

 

      your recommendation?

 

                DR. COONEY:  So, the request is to embrace

 

      the content of slides two, three and four?

 

                DR. HUSSAIN:  Yes.

 

                DR. COONEY:  The procedure that we should

 

      follow for a vote--we will go around the table and

 

      we will begin with Art and I will fill in the last

 

      step.  You have three options, yes, no or abstain.

 

                DR. KIBBE:  Never give me three choices;

 

      it is too hard on me.  Yes.

 

                DR. COONEY:  Marv?

 

                DR. MEYER:  Yes.

 

                DR. SINGPURWALLA:  Yes.

 

                DR. COONEY:  Carol?

                                                               283

 

                DR. GLOFF:  I am supposed to vote?

 

                DR. COONEY:  Yes.

 

                DR. GLOFF:  Yes.

 

                DR. DELUCA:  Yes.

 

                DR. MORRIS:  Yes.

 

                DR. COONEY:  Mike?

 

                DR. KORCZYNSKI:  Yes.

 

                DR. SWADENER:  Yes.

 

                DR. COONEY:  Cynthia?

 

                DR. SELASSIE:  Yes.

 

                DR. COONEY:  Tom?

 

                DR. LAYLOFF:  Yes.

 

                DR. COONEY:  My vote is also yes.

 

      Therefore, we have 11 yes, zero no and zero

 

      abstentions for the vote.  Wonderful!  Thank you

 

      all for your input and your discussion.  The next

 

      piece will be by video conference or

 

      teleconference.  It is scheduled for 4:30.  We are

 

      hopeful that we can get Jurgen on the line before

                                                               284

 

      that.  But would anybody object if I gave you a

 

      ten-minute break?  Ken has to stay here and work, a

 

      10-minute break for everyone else while we set up

 

      the teleconference.

 

                [Brief recess]

 

               Clinical Pharmacology Subcommittee Report

 

                          (via teleconference)

 

                DR. COONEY:  We are about to begin the

 

      final part of today's program.  Jurgen Venitz I

 

      think is on line.

 

                DR. VENITZ:  Yes, I am here.

 

                DR. COONEY:  I believe that you can hear

 

      everything that is said into a microphone.

 

                DR. VENITZ:  That is correct.

 

                DR. COONEY:  So, if anyone wishes to speak

 

      please turn your microphone on.  I also understand

 

      that we have a set of slides with your name on

 

      them.  They are showing up on the screen, and I

 

      would invite you to begin.

 

                DR. VENITZ:  Thank you for giving me the

 

      opportunity to discuss on behalf of the Clinical

 

      Pharmacology Subcommittee what transpired at our

                                                               285

 

      last meeting.

 

                As you can tell from the first slide, this

 

      committee provides expertise to the parent Advisory

 

      Committee for Pharmaceutical Science on

 

      exposure-response modeling, pediatric clinical

 

      pharmacology and pharmacogenetics.

 

                The next slide shows you the topics which

 

      were discussed at our most recent meeting in

 

      November of last year.  We had five topics being

 

      discussed, one being an update on the progress the

 

      subcommittee had made.  Then we spent a

 

      considerable amount of time on pharmacogenetic

 

      testing of irinotecan, followed by a discussion on

 

      drug-drug interaction potential assessment; a

 

      tribute to Lewis Sheiner and, on the second day

 

      discussed the role of biomarkers and surrogate

 

      markers.

 

                As you can tell, those slides are more

 

      explicit than my usual summary, the reason being

 

      that... [speaker phone problems;

 

      inaudible]...exposure-response and simulation

 

      guidances where the committee was having an impact

                                                               286

 

      were used internally within the OCPB and continue

 

      to have an impact on labeling recommendations.  He

 

      also told us that discussion

 

      of...[inaudible]...risk assessment that had been

 

      going on for about two years were on hold, the

 

      reason being there was no consensus internally and

 

      externally as to how to come up with an acceptable

 

      way for setting up ...[inaudible].

 

                On the next slide he also shared with us

 

      that the [inaudible]...decision tree was continued

 

      to be used within FDA and there was a research

 

      project going on within OCPB that was trying to

 

      assess different [inaudible]...in children and how

 

      to assess and revise the pediatric decision tree.

 

                The next slide... [inaudible]...in using

 

      pharmacogenetic testing for TMP, an enzyme involved

 

      in the metabolism of...[inaudible]...labeling

 

      language to include that testing in 2003 and at the

 

      time, meaning as of end of 2004, negotiations

 

      between the FDA and the sponsors were ongoing to

 

      analyze labeling language that would at least

 

      recommend a test for TMPT.  [Inaudible].

 

                On the next slide, Dr. Lesko shared with

 

      us that at the end-of-phase 2a meeting initiative

 

      that he undertook late in 2003 was ongoing and that

                                                               287

 

      a guidance was being developed and should be coming

 

      out this year.

 

                Lastly he told us that a QT-liability

 

      discussion that has been going

 

      on...[inaudible]...discussions were going on within

 

      the agency to...[inaudible]...clinical study

 

      designs and analyses.

 

                The second part, as I mentioned before was

 

      the role of pharmacogenetic testing for irinotecan.

 

      [Inaudible]...Rahman, Dr. Parodi with respect to

 

      Pfizer, and Dr. Ratan was invited as the consultant

 

      to discuss...[inaudible].

 

                Irinotecan is an oncological agent.  It is

 

      currently approved and used for first-and

 

      second-line treatment of colorectal cancer.  Like

 

      most cancer drugs, it is limited in its use by

 

      major clinical toxicities, primarily neutropenia

 

      that cases infection and diarrhea.  On the other

 

      hand, CPT-11 has...[inaudible]...pharmacokinetics. 

                                                               288

 

      The drug is just a

 

      prodrug...[Inaudible]...metabolized by SN-38.  This

 

      metabolite, SN-38 is further metabolized by an

 

      enzyme called UGT1A1, which forms inactive

 

      glucuronide.  The peculiarity about this enzyme,

 

      UGT1A1, is that it is subject to

 

      pharmacokinetic...[inaudible]...has an allele

 

      called 7/7 that is prevalent in...[inaudible]...to

 

      form glucuronide. However, there are other enzymes,

 

      such as CYP3A4 and other transporters such as P-gp

 

      that are involved in irinotecan PK as well.  Their

 

      significance clinically is unknown as yet.

 

                What was known at the time of the meeting

 

      was that SN-38 as the active metabolite was

 

      associated with the *28 genotype.  So,

 

      patients...[inaudible]...also have higher exposure

 

      of active metabolite, which is what you would

 

      expect.  Furthermore, systemic experience to this

 

      metabolite...[inaudible]...namely, neutropenia.

 

      The risk of grade 4 neutropenia was felt to be 9.3

 

      for patients that have 7/7 genotype.  So,

 

      patients...[inaudible]...to develop very severe

                                                               289

 

      neutropenia.  The second limiting toxicity,

 

      diarrhea, was less clearly associated with the

 

      genotype.                [Inaudible]...label irinotecan

 

      with certain known risk

 

      factors...[inaudible]...responsible for

 

      neutropenia, in particular, age, prior

 

      abdominal/pelvic radiation, low performance status

 

      and increased bilirubin.  So, the discussion or the

 

      vote the committee was asked to take was can we use

 

      UGT1A1 genotype as a risk factor that would then

 

      lead to a relabeling of...[inaudible].  So, we were

 

      presented with a summary of clinical studies that

 

      dealt with that issue.  So, across four clinical

 

      trials--the information on the next slide shows you

 

      that the odds ratio for patients that have 7/7 gene

 

      variant--the odds for neutropenia were raised from

 

      2.5 to 16.7.  So, obviously those patients were at

 

      significantly increased risk of developing

 

      neutropenia. On the other hand, diarrhea, as I

 

      said, was much less clearly associated with this

 

      genotype.

 

                The limitations for all those studies were

                                                               290

 

      the fact that they were not designed to assess the

 

      strength of the genetic association.  Nevertheless,

 

      Pfizer presented the committee, on the next slide,

 

      with a table that gives you the estimated

 

      performance battery for this test.  The sensitivity

 

      of this test in those four clinical studies would

 

      be 22 percent, however, the specificity would

 

      expected to be 95 percent.  This means that you

 

      would expect negative fixed value of 83 percent.

 

      This really means that if you take somebody and

 

      somebody does have this allele there is an 83

 

      percent chance that this individual would not

 

      develop neutropenia.  On the other

 

      hand...[inaudible]...would be a patient that has

 

      this neutropenia and does have the genotype.  There

 

      is a 50 percent chance of that.  This is shown as

 

      an overall incidence of neutropenia of

 

      about...[inaudible].

 

                This was further elaborated on

 

      by...[inaudible] ...and he shared with the

 

      committee that in his estimation without

 

      pharmacogenetic testing 100 percent of the patients

                                                               291

 

      were treated and 10 percent of those developed

 

      severe neutropenia.  On the other hand, if this

 

      test is implemented, only 90 percent of patients

 

      treated only 5 percent would develop severe

 

      neutropenia.  So, the test

 

      actually...[inaudible]...many patients would be

 

      tested in order to protect...[inaudible].

 

                Now, there were some concerns raised

 

      during the discussion about a role and implications

 

      of...[inaudible].  The first was that current

 

      studies were really limited in assessing the

 

      strength of association between the negative

 

      clinical outcome of neutropenia and the UGT1A1 *28

 

      genotype. Pfizer also indicated to us that ongoing

 

      clinical trials may help identify other variants,

 

      other than UGT

 

      ...[inaudible].  Currently, there is no validated

 

      algorithm for dosing that would allow us to adjust

 

      the doses after PG testing has been performed.

 

      And, there was some concern that if you reduced

 

      irinotecan...[inaudible].

 

                The committee was asked to vote. 

                                                               292

 

      [Inaudible]... that this 7/7 genotype is associated

 

      with a higher risk of neutropenia and, as you can

 

      tell, the committee unanimously agreed with that.

 

      The second statement or the second question put to

 

      us was that 7/7 genotype was associated with a

 

      higher risk for acute or delayed diarrhea.  The

 

      committee didn't believe that there was sufficient

 

      evidence to support that.  The most important vote

 

      was the last where we were asked does *28 PG

 

      testing have adequate sensitivity and specificity,

 

      and the committee voted 9 in favor and 3

 

      abstentions.

 

                During the discussion there were some

 

      comments by the committee to include PG testing in

 

      the label even though...[inaudible].  We noted that

 

      there is lack of information in the current label

 

      about the dosing regimen, which is not unusual.  We

 

      were...[inaudible]...that lower doses as a result

 

      of the test may actually allow patients to stay on

 

      drug...[inaudible]...as opposed to discontinuing

 

      treatment because of neutropenia.  We realized,

 

      however, that additional clinical testing may be

                                                               293

 

      needed to establish and validate a modified dosing

 

      regimen.  And there was some discussion about

 

      bilirubin which, as I mentioned before is a UGT1A1

 

      substrate, as a potential safety marker in addition

 

      to the pharmacogenetic testing.

 

                The second topic for discussion on the

 

      first day dealt with the issue about drug

 

      interactions.  You can tell

 

      ...[inaudible]...starting with Dr. Huang, followed

 

      by Gottesdiener, Lacluyse and Reynolds from OCPB as

 

      well.  The issues here were around the update of

 

      the DDI guidance.  In particular, we were

 

      asked...[inaudible]...in vitro transfer studies;

 

      how to integrate in vitro enzyme induction studies,

 

      and whether there should be a requirement for

 

      inhibitor/impairment in vivo studies.

 

      [Inaudible]...and also eliminated by the kidney.

 

      Then the question is should you study this drug in

 

      patients who have...[inaudible]...as well as have a

 

      genetic variant of 2D6 enzyme that makes them poor

 

      metabolizers.

 

                You can see in the next slide that we were

                                                               294

 

      basically...[inaudible]...and the committee was

 

      asked to vote in favor or against.

 

      [Inaudible]...that has shown in vitro not to have

 

      inhibition of 1A2, 2C9, 2C19 and 2D6 and 3A.  Is

 

      there a need for an in vitro study?  And, the

 

      committee pretty much unanimously agreed that there

 

      wasn't any need to do an in vivo study.  So, in

 

      vitro would predict the in vivo.

 

                The next question, if you have an NME that

 

      is a P-gp inhibitor in vitro should there be a

 

      requirement or should there be a follow-up in vivo

 

      interaction study with a known P-gp substance?

 

      Again by majority the committee was in favor of

 

      doing an in vivo study as a result of in vitro

 

      demonstrated inhibition.

 

                The next question was a little more

 

      complicated.  So, here the NME, the new molecular

 

      entity, is a P-gp substrate and a 3A4 substrate,

 

      and the question was is there a need to do an in

 

      vivo interaction study with a drug like ritonavir

 

      which inhibits both P-gp as well as 3A4, and again,

 

      by a smaller margin, the committee voted in favor

                                                               295

 

      of that.

 

                The next question related to an NME that

 

      is a P-gp substrate and not a 3A4 substrate.

 

      Should there be an in vivo study specifically to

 

      look at P-gp inhibition and the committee, with a

 

      very slim margin, voted in favor.

 

                Then we moved on to new cytochrome P54

 

      enzyme tests.  The question was put to us would the

 

      committee recommend in vivo drug interaction

 

      studies for CYP2B6, 2B8 and for this UGT1A1 enzyme,

 

      and the committee was virtually unanimous in favor.

 

      then we moved along to look at transporter systems,

 

      such as OATP and MRP, and we were asked would we

 

      recommend in vivo drug interaction studies and, as

 

      you can tell, the majority of the committee was

 

      opposed to that, the main reason being that we

 

      didn't think that science would really allow us to

 

      draw any mechanistic conclusions from in vivo drug

 

      interaction studies.

 

                Then we moved to in vitro induction and

 

      its predictive value.  We were asked if an in vitro

 

      induction study for a new molecular entity has more

                                                               296

 

      than 40 percent ...[inaudible]...positive control

 

      should an in vivo study be done.  The committee, by

 

      majority, voted against that.

 

                The next question was if there was in

 

      vitro demonstration of lack of that, there is no

 

      need for any in vivo studies...[inaudible], and the

 

      committee felt evidence exists currently

 

      so...[inaudible]...by majority.

 

      [Inaudible]...should inhibitor in vivo studies be

 

      recommended to actually do that and the committee,

 

      by virtually unanimous vote, voted against it.

 

                Then we moved to the second day of our

 

      meeting.  That was started with a tribute to Dr.

 

      Sheiner. That was given by Blaschka who is at

 

      University of California at San Francisco.  Lew

 

      Sheiner, as some of you may

 

      know...[inaudible]...worked as a consultant for a

 

      long time.  He was known as a seminal researcher

 

      and teacher in the area of PK/PD exposure-response

 

      and pharmacometrics, and a lot of terms and

 

      approaches that he developed still are being used

 

      quite extensively--learn and confirm cycles in drug

                                                               297

 

      development; the role of empiricism versus

 

      mechanistic approaches, and the issue of

 

      frequentists versus Bayesian statisticians.  He

 

      also developed what is called "Sheiner's rules"

 

      that deal with the certainty of knowledge that is

 

      used or required.

 

                [Inaudible]...the role of biomarkers,

 

      surrogate markers and regulatory decision-making.

 

      These presentations, starting with Dr. Woodcock of

 

      CDER, Dr. Wagner, from Merck on behalf of PhRMA,

 

      and Dr. Blaschke.

 

                Dr. Woodcock led a discussion, and I guess

 

      it was more a...[inaudible]...than it was a

 

      discussion but she shared with us biomarkers

 

      indicate biological processes and/or the

 

      pharmacological responses to therapeutic

 

      intervention.  So, anything that changes as a

 

      result of these or the drug is considered to be a

 

      biomarker.  On the other hand, clinical endpoints

 

      measure how patients feel, function or survive and

 

      are related to outcomes such as efficacy and/or

 

      this of a drug.  [Inaudible]...somewhat in between.

                                                               298

 

      So, those can be measured and are intended to

 

      replace clinical endpoints for efficacy and

 

      toxicity.

 

                Dr. Woodcock shared with us in her talk

 

      that the rational use of those biomarkers can,

 

      indeed, accelerate the drug development process and

 

      the internal decision-making

 

      within...[inaudible]...as well as regulatory

 

      decision-making within the FDA.  She also pointed

 

      out that biomarkers can provide a mechanistic

 

      bridge between preclinical studies that is

 

      typically...[inaudible]...and the clinical test

 

      which is typically quite empiric.  However, in

 

      order for this to really have an impact, a new

 

      business model needs to be developed that allows

 

      biomarkers to be developed in parallel with drug

 

      development as part of a commercial enterprise.

 

                [Inaudible]...and are rarely met in a

 

      strict statistical sense.  She pointed out future

 

      clinical endpoints may not be a univariate as we

 

      currently use them but...[inaudible]...and that

 

      biomarkers may help get away from a mean analysis

                                                               299

 

      to a responder analysis... [inaudible]...pave the

 

      way for this individualization and personalization

 

      of...[inaudible]...in clinical testing as well as

 

      post-market.

 

                Her presentation was followed by Dr.

 

      Blaschke who did review use of HIV viral load

 

      as...[inaudible].  He pointed out the validation of

 

      those surrogate markers required sensitive assays

 

      to detect...[inaudible]...as well as mechanistic

 

      models about disease progression, both qualitative

 

      as well as intellectual.

 

                He pointed out that biomarkers need to be

 

      causal path.  That means they have to be

 

      mechanistically... [inaudible] and proximal to the

 

      disease endpoint in order to provide confirmatory

 

      evidence to support the efficacy of new molecular

 

      entities.

 

                The last presentation was Dr. Wagner's

 

      presentation which, as I mentioned before, spoke on

 

      behalf of the work group that PhRMA put together do

 

      deal with this issue of biomarkers and surrogate

 

      markers.  He reviewed what is called

                                                               300

 

      "fit-for-purpose" qualification of biomarkers,

 

      meaning the extent of qualification of a biomarkers

 

      depends on its intended use.  He reviewed four uses

 

      for it.  The first one would be the exploratory

 

      use.  So, here the biomarker is used as a research

 

      tool and, obviously, the requirements in terms of

 

      qualification are relatively minor.

 

                The next level of qualification would be a

 

      demonstration of purposes  So, those would be

 

      biomarkers that are likely or emerging as useful

 

      biomarkers.

 

                The next level would be characterization.

 

      So, here you are using known or established

 

      biomarkers to assess exposure-response

 

      relationships and mechanism of action, for example.

 

                The highest level of qualification would

 

      obviously be surrogacy so here the biomarkers would

 

      become a surrogate marker and would substitute for

 

      clinical endpoints.  That would obviously require

 

      the highest level of qualification.

 

                Dr. Wagner did lament the lack of

 

      nomenclature in the biomarker area.  He spoke of

                                                               301

 

      the need for collaboration between PhARMA, FDA, NIH

 

      and academia on two issues, number on, what are the

 

      most useful biomarkers to pursue that might, in the

 

      long run, provide a payoff and, more importantly,

 

      how to decide what evidence can be used and how it

 

      can be used to accept biomarkers and surrogate

 

      markers as part of a regulatory...[inaudible], and

 

      he also mentioned some of the hurdles associated

 

      with extensive use of biomarkers such as

 

      incentives, intellectual property rights and

 

      funding.

 

                [Inaudible]...that the discussion of

 

      markers would continue in one of the next meetings.

 

      And, that is all I have to report.

 

                     Questions by Committee members

 

                DR. COONEY:  Thank you very much.  We have

 

      an opportunity for questions from the committee.

 

      Ajaz?

 

                DR. HUSSAIN:  Jurgen, thanks for reporting

 

      the subcommittee report.  I think Prof. Nozer left

 

      before you mentioned the Bayesian aspect so you

 

      missed that.

 

                One question I had was I think in the work

 

      we are doing also from a risk-based perspective

 

      utility functions really have to be discussed and

                                                               302

 

      we need to start thinking about that.  What are the

 

      challenges that you are facing that has started

 

      that discussion?

 

                DR. VENITZ:  Well...[inaudible]...there

 

      was no consensus on not only how to come up with a

 

      uniform rating scale but also a process to follow

 

      because you have to be involve all

 

      stakeholders--patients, clinicians, sponsors,

 

      regulatory individuals--in designing a process

 

      before you can get utility...[inaudible]...results.

 

      So, that is the reason why he told us that for the

 

      time being that initiative has been put on hold.  I

 

      didn't get the sense that it was fatally wounded as

 

      much as they were trying to reassess what to do

 

      internally.

 

                DR. HUSSAIN:  One more comment that I

 

      have, Jurgen, usually you are here but I think one

 

      opportunity you have is if you really looked at the

 

      PAT guidance and what you are doing in clinical

                                                               303

 

      pharmacology has a lot of commonality, and the

 

      biomarkers, surrogacy and so forth I think comes to

 

      the same level of discussion in terms of

 

      fundamentals to alternate testing and control

 

      strategies that we are developing.  So, keep that

 

      in mind, and maybe have some discussion on the

 

      commonality and how we might approach things that

 

      might be useful at some future point.

 

                DR. VENITZ:  I would agree with that and I

 

      would add, as was mentioned in the tribute that was

 

      given to Dr. Sheiner, his approach, which I think

 

      is very pertinent to the PAT approach as well, is

 

      how much weight do you give empiric evidence by

 

      testing and testing over again, and how much do you

 

      give to mechanistic understanding whether it is a

 

      manufacturing process or whether it is the

 

      pathogenicity of disease.  That is really what the

 

      dilemma is.  As I mentioned before, the

 

      frequentists would just do things over and over

 

      again if you are willing to make any mechanistic

 

      assumptions.  Of, if you are more on the

 

      mechanistic side you might be willing to make

                                                               304

 

      certain assumptions without having to empirically

 

      repeat experiments and just base your confidence on

 

      the reproducibility of the test.

 

                DR. HUSSAIN:  Thank you.

 

                DR. COONEY:  Thank you.  Jurgen, the good

 

      news and the bad news at the end of the day is that

 

      it is the end of the day.  Cynthia?

 

                DR. SELASSIE:  Jurgen, I have a question

 

      for you.  Do increased levels of P-gp result in

 

      greater levels of neutropenia?  Does it impact it

 

      in any way?

 

                DR VENITZ:  As far as I know, there are no

 

      clinical studies.  All we know is that G-pg seems

 

      to be involved in the kinetics of irinotecan.

 

      Whether... [inaudible] is a very difficult question

 

      at this stage to answer.  The only thing we know is

 

      that UGT1A1 has been shown to be associated with

 

      neutropenia.  None of those tests have proven to be

 

      clinically relevant...[inaudible]... any

 

      association between neutropenia levels of

 

      irinotecan and those genetic...[inaudible].  So,

 

      right now the answer is we do not know.

 

                DR. COONEY:  Thank you.  I think we have

 

      reached the end of our discussion, looking at the

 

      way the people are sitting around the room.  Thank

                                                               305

 

      you very much for the summary and the update.

 

                DR. VENITZ:  Good luck.  I am sorry that I

 

      couldn't be there.

 

                DR. COONEY:  Thank you.  We will look

 

      forward to you joining us next time.

 

                DR. VENITZ:  Okay.  Bye, bye.

 

                DR. COONEY:  I think we can adjourn the

 

      meeting unless anyone else has anything pressing.

 

      Everyone looks like they are ready to adjourn!

 

      Thank you all very much.  I look forward to seeing

 

      you tomorrow morning.

 

                [Whereupon, the proceedings were adjourned

 

      at 4:42 p.m., the reconvene at 8:30 a.m.,

 

      Wednesday, May 4, 2005.]

 

                                 - - -