1

 

                DEPARTMENT OF HEALTH AND HUMAN SERVICES

 

                      FOOD AND DRUG ADMINISTRATION

 

                CENTER FOR DRUG EVALUATION AND RESEARCH

 

 

 

 

 

 

 

 

 

 

 

             ADVISORY COMMITTEE FOR PHARMACEUTICAL SCIENCE

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

                      Wednesday, October 20, 2004

 

                               8:30 a.m.

 

 

 

 

 

 

 

 

 

                CDER Advisory Committee Conference Room

                           5630 Fishers Lane

                          Rockville, Maryland

                                                                 2

 

                              PARTICIPANTS

 

      Arthur H. Kibbe, Ph.D., Chair

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

 

      MEMBERS

      Patrick P. DeLuca, Ph.D.

      Paul H. Fackler, Ph.D.

      Meryl H. Karol, Ph.D.

      Melvin V. Koch, Ph.D.

      Michael S. Korczynski, Ph.D.

      Marvin C. Meyer, Ph.D.

      Gerald P. Migliaccio, Ph.D. (Industry

      Representative)

      Kenneth R. Morris, Ph.D.

      Cynthia R.D. Selassie, Ph.D.

      Nozer Singpurwalla, Ph.D.

      Marc Swadener, Ed.D. (Consumer Representative)

      Jurgen Venitz, M.D., Ph.D.

 

      SPECIAL GOVERNMENT EMPLOYEES SPEAKERS

      Judy Boehlert, Ph.D.

      Gordon Amidon, Ph.D., M.A.

      FDA Staff

      Gary Buehler, R.Ph.

      Lucinda Buhse, Ph.D.

      Jon Clark, M.S.

      Jerry Collins, Ph.D.

      Joseph Contrera, Ph.D.

      Ajaz Hussain, Ph.D.

      Monsoor Khan, R.Ph., Ph.D.

      Steven Kozlowski, M.D.

      Vincent Lee, Ph.D.

      Qian Li, Ph.D.

      Robert Lionberger, Ph.D.

      Robert O'Neill, Ph.D.

      Amy Rosenberg, M.D.

      John Simmons, Ph.D.

      Keith Webber, Ph.D.

      Helen Winkle

      Lawrence Yu, Ph.D.

                                                                 3

 

                            C O N T E N T S

                                                              PAGE

 

      Call to Order

        Arthur Kibbe, Ph.D.                                      5

 

      Conflict of Interest Statement:

        Hilda Scharen, M.S.                                      5

 

      Committee Discussion (Continued)                           8

 

      Science in Regulation - Visionary Overview

        Arthur Kibbe, Ph.D.                                     43

 

      The "Desired State" of Science-and Risk-based

        Regulatory Policies

          Ajaz Hussain, Ph.D.                                   74

 

        Organization Gap Analysis

          Helen Winkle                                          75

 

        Scientific Gap Analysis

          Ajaz Hussain, Ph.D.                                  103

 

        Policy Gap Analysis

          Jon Clark, M.S.                                      135

 

      Generic Pharmaceutical Association Perspective

        Shahid Ahmed, M.S.                                     152

 

      Pharmaceutical Research and Manufacturers

      of America Perspective

        Gerry Migliaccio, Ph.D.                                164

 

      Committee Discussion and Recommendations                 182

 

      Pharmaceutical Equivalence and Bioequivalence

      of Generic Drugs

 

        The Concept and Criteria of BioINequivalence

        Concept of BioINequivalence

          Lawrence Yu, Ph.D.                                   205

 

        Criteria of BioINequivalence

          Qian Li, Sc.D.                                       222

                                                                 4

 

                      C O N T E N T S (Continued)

 

                                                              PAGE

 

      Committee Discussion and Recommendations                 234

 

      Bioequivalence Testing for Locally Acting

      Gastrointestinal Drugs

 

        Topic Introduction

          Lawrence Yu, Ph.D.                                   273

 

        Scientific Principles

          Gordon Amidon, Ph.D.                                 275

 

        Regulatory Implications and Case Studies

          Robert Lionberger, Ph.D.                             308

 

      Committee Discussion and Recommendations                 324

 

      Conclusion and Summary Remarks

        Ajaz Hussain, Ph.D.                                    341

        Helen Winkle                                           349

 

                                                                 5

 

                         P R O C E E D I N G S

 

                             Call to Order

 

                DR. KIBBE:  Ladies and gentlemen, I would

 

      like to call the meeting to order.  The first item

 

      of business is the reading of the Conflict of

 

      Interest Statement.

 

                     Conflict of Interest Statement

 

                MS. SCHAREN:  Good morning.  The following

 

      announcement addresses the issue of conflict of

 

      interest with respect to this meeting and is made a

 

      part of the record to preclude even the appearance

 

      of such.

 

                Based on the agenda, it has been

 

      determined that the topics of today's meeting are

 

      issues of broad applicability and there are no

 

      products being approved.  Unlike issues before a

 

      committee in which a particular product is

 

      discussed, issues of broader applicability involve

 

      many industrial sponsors and academic institutions.

 

                All Special Government Employees have been

 

      screened for their financial interests as they may

 

      apply to the general topics at hand.  To determine

 

                                                                 6

 

      if any conflict of interest existed, the Agency has

 

      reviewed the agenda and all relevant financial

 

      interests reported by the meeting participants.

 

                The Food and Drug Administration has

 

      granted general matters waivers to the Special

 

      Government Employees participating in this meeting

 

      who require a waiver under Title 18, United States

 

      Code, Section 208.

 

                A copy of the waiver statements may be

 

      obtained by submitting a written request to the

 

      Agency's Freedom of Information Office, Room 12A-30

 

      of the Parklawn Building.

 

                Because general topics impact so many

 

      entities, it is not practical to recite all

 

      potential conflicts of interest as they apply to

 

      each member, consultant, and guest speaker.

 

                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

 

      representative, we would like to disclose that Dr.

 

                                                                 7

 

      Paul Fackler and Mr. Gerald Migliaccio are

 

      participating in this meeting as non-voting

 

      industry representatives acting on behalf of

 

      regulated industry.  Dr. Fackler's and Mr.

 

      Migliaccio's role on this committee is to represent

 

      industry interests in general, and not any other

 

      particular company.

 

                Dr. Fackler is employed by Teva

 

      Pharmaceuticals U.S.A., and Mr. Migliaccio is

 

      employed by Pfizer, Inc.

 

                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 participants' involvement and their

 

      exclusion will be noted for the record.

 

                With respect to all other participants, we

 

      ask 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. KIBBE:  Thank you.

 

                                                                 8

 

                Yesterday, we concluded with a suggestion

 

      that we might want to continue our discussion about

 

      the questions that the Agency has raised, and I

 

      think Dr. Meyer has asked Dr. Hussain to come up

 

      with a straw man, and we have it ready, so I think

 

      we should go there first and then go back to the

 

      scheduled agenda.

 

                Ajaz.

 

                    Committee Discussion (Continued)

 

                DR. HUSSAIN:  Good morning.  I think the

 

      discussions towards the end of yesterday started

 

      honing down on some of the key challenges we face

 

      in the designing of a Critical Path Initiative in

 

      OPS.

 

                I think, reflecting back on the discussion

 

      yesterday, clearly, I think we have a wide range of

 

      research capabilities and programs already in

 

      place, and the challenge would be to sort of direct

 

      these in a very focused way to help the Critical

 

      Path Initiative, keeping in mind that all of our

 

      research will not be focused on critical path,

 

      there are other aspects that we have to focus on.

 

                                                                 9

 

                I will sort of reflect back on the PAT

 

      Initiative and how that sort of evolved.  Clearly,

 

      if you recall, the PAT Initiative led to the GMP,

 

      and there is a whole sequence of initiatives that

 

      have occurred.  The PAT Initiative was a model and

 

      we can learn some things from that as a model also.

 

                I will sort of summarize my thoughts here

 

      with a hypothesis statement that Jerry proposed

 

      yesterday, that Critical Path Initiative will

 

      improve the efficiency and effectiveness of drug

 

      development process.  That is the hypothesis that

 

      sort of really we are engaged in trying to fulfill

 

      or trying to confirm.

 

                The challenge would be then how do we

 

      measure efficiency and effectiveness of drug

 

      development.  That is one of the keys, how do you

 

      measure drug development in terms of the failure

 

      rate, or the time it takes, or the cost of drug

 

      development.

 

                All of these are relevant metrics, but for

 

      the purposes of a hypothesis, what and how should

 

      we approach and define that, because unless you can

 

                                                                10

 

      measure something, you cannot improve it.  So,

 

      measurement and metrics would be a key factor of

 

      that.

 

                The second aspect then would be what are

 

      the root causes of low efficiency and effectiveness

 

      in all the three dimensions.  A number of factors

 

      were put up looking at 1999 or 1991 or 2000, and so

 

      forth.

 

                They were indicators of what may be

 

      happening, but you have to keep in mind that is

 

      partial information, information available to FDA

 

      and available in public is just limited because the

 

      companies have far more information about the root

 

      causes, and so forth.  So, you have to sort of

 

      factor that into our decisionmaking.

 

                The next question is who is in the best

 

      position to address these root cause factors that

 

      we identify, what is the role of FDA, what should

 

      FDA do and what should some industry, academia, and

 

      other agencies should be doing is the key there.

 

                I think based on information and based on

 

      experience at FDA, clearly, we are in a good

 

                                                                11

 

      position to identify many of the problems, not all

 

      of the problems, but many of the problems, and FDA

 

      has the responsibility to communicate these

 

      findings in some way or form.

 

                If you look at John Simmons' presentation,

 

      he laid out, as a part of the critical path,

 

      strategic meeting points during the drug

 

      development process.  That is one aspect,

 

      communication between sponsors of applications and

 

      our review scientists, if that is timely and in a

 

      coordinated manner, that is one effective means of

 

      that.

 

                So, communication through meetings for

 

      specific drug applications, broader communications

 

      with workshops, and then eventually guidance

 

      documents outlining FDA's current thinking on a

 

      given topic are the communication mechanisms that

 

      we have.

 

                For that, clearly, I think you have to

 

      think about resources and how do you facilitate

 

      that process.  If you want to sort of move towards

 

      more meetings and more interactions between

 

                                                                12

 

      reviewers and sponsors, then, you have to build the

 

      time for that, and so forth.  That has to be

 

      considered, too.

 

                Workshops and guidances also take a

 

      significant amount of effort, and so forth, so as

 

      we improve our communication channels, where will

 

      we find the resources and  time to do that, I think

 

      that is also a management aspect that has to be

 

      discussed.

 

                FDA's knowledge base, I think was clearly

 

      an asset in the sense we have a lot of information.

 

      If we are able to create a knowledge base that can

 

      be useful, not only for identifying problems that

 

      we see, but also for improving of a predictive

 

      ability in all three dimensions, safety, efficacy,

 

      and industrializations, what are the practices that

 

      lead to success, what are the practices that may

 

      not be as efficient, and so forth.

 

                So, this knowledge base would be useful

 

      for that purpose, but again I will remind in the

 

      sense we have to be cautious, there are limitations

 

      of that knowledge, because we don't always have all

 

                                                                13

 

      the information, so you have to factor that in.

 

                But based on our knowledge base and based

 

      on communication, and so forth, I think the

 

      laboratory and the research functions clearly have

 

      to focus on improving methodologies.  There are

 

      many aspects of laboratory work that only FDA is in

 

      a good position to do, and others either don't have

 

      the interest or don't have the focus to sort of

 

      address some of the challenges.

 

                For example, in the case of regulatory

 

      decisionmaking, risk-based decisionmaking, the

 

      decision process itself often needs support of

 

      science, and so forth, so that is where the

 

      research really could focus on.

 

                Also for development and validation of new

 

      methodologies, standards development, methodology,

 

      validation, say, from biomarker to any new

 

      technology, unless FDA has a role in achieving

 

      that, it may not be fully appreciated within the

 

      Agency, and some of the Agency concerns would not

 

      be addressed if it is done totally outside, so

 

      there has to be some means of linking our

 

                                                                14

 

      laboratory work to standards development,

 

      validation of new methods, and so forth.

 

                Our postmarketing experience again is

 

      unique because that is where I think we have a lot

 

      of information, how do we capture that as lessons

 

      learned and how do we use that.  You saw some

 

      examples of how we were learning from that and

 

      going retrospectively and said how could we have

 

      improved the process.  Those experiments would be

 

      very valuable.

 

                One aspect is in terms of innovation, in

 

      terms of new technologies, what is an important

 

      aspect of standard setting?  Standard setting and

 

      guidances are slightly different in my opinion.

 

      For example, in the PAT Initiative, we opted to

 

      move towards ASTM International as the body for

 

      standard setting.

 

                What that does is allows industry,

 

      academia, every stakeholder to be part of that, and

 

      actually identify what standards are needed, and

 

      actually develop those as quickly as possible.

 

                That relieves the burden on FDA, and FDA

 

                                                                15

 

      simply adapt or adopt those standards after

 

      evaluation, so that might be an option that seems

 

      to be moving forward in the PAT Initiative for new

 

      technologies, new methods, and so forth.  So, that

 

      could be considered at the same time.

 

                But at the same time, I think as we look

 

      at FDA's role, what is the role of industry and

 

      what is the role of other agencies and academia

 

      really have to sort of come together.

 

                The role of industry I think is knowledge

 

      sharing. Clearly, it has far more information, and

 

      reluctance to share knowledge will inhibit the

 

      progress, and how do you do that is a key

 

      challenge.

 

                At the same time, I think in order to

 

      bring all of us together, focused on a given goal,

 

      we really I think have to define clearly the

 

      metrics, the desired state, and so forth, and come

 

      on the same page, so that we can coordinate all of

 

      these activities.

 

                In some ways, FDA could play that role of

 

      coordination, as Viad declared yesterday, not only

 

                                                                16

 

      from the prospective of we are not competing in

 

      this arena, eventually, we have to be involved, so

 

      coordination function for FDA would be an important

 

      function for all these activities.

 

                If we have a clear understanding of what

 

      are the issues and what are we trying to achieve,

 

      then, the coordination and synergy would sort of

 

      evolve naturally.

 

                So, those are the sort of thought process

 

      that I could capture.

 

                DR. KIBBE:  Anybody?  Marvin?  You asked

 

      for the straw, you got the straw man.

 

                DR. MEYER:  The virus, are you talking

 

      about that later?

 

                DR. HUSSAIN:  No, that is a very specific

 

      example.

 

                DR. MEYER:  I thought I had more time to

 

      think then, since I was waiting for the virus.

 

                DR. KIBBE:  Does somebody else want to--

 

                DR. MEYER:  No, no, I have something to

 

      say.

 

                DR. KOCH:  Marvin, just before you--I

 

                                                                17

 

      think I need a point of clarification because some

 

      of what came out yesterday was the desire to have

 

      either shorter development time, more compounds

 

      coming out that could be effective, new

 

      pharmaceuticals, and it seems to be a push towards

 

      industry to try to become more effective, et

 

      cetera, but I saw a couple times yesterday where

 

      things developed within the Agency to improve the

 

      ability to go after materials through some of the

 

      databases and things were certainly ways to help

 

      that process.

 

                The other thing, though, is that when you

 

      looked at that chart that showed an increase in

 

      cost of materials and a few other things, toxicity,

 

      you know, is something that shows up there, and I

 

      think something a little bit insidious over time

 

      has been with improved technologies and increased

 

      concerns over pharmaceuticals, there are new tests

 

      that come in that prolong the evaluation, that

 

      anything the Agency can do to pull things together

 

      to make those things, immunogenicity or other

 

      things that have, you know, you go back two

 

                                                                18

 

      generations ago and if you come up with a new

 

      material, would you put it through all of the same

 

      tests, so anything that can be done to simplify and

 

      do more predictive studies in that regard, I think

 

      would help.

 

                DR. HUSSAIN:  Definitely, that is an

 

      important point.  For example, I think as we move

 

      towards more complex materials, the material cost

 

      is, as you saw, is already showing up, and so

 

      forth.

 

                Introduction of new excipients or new

 

      adjuvants, and so forth, is a significant

 

      challenge, and as we go towards nanomaterials,

 

      nanodevices, and so forth, if we still have to rely

 

      on the traditional pharmaceutical excipients, it

 

      would be a very limiting aspect, so I think that

 

      that is clearly on our agenda.

 

                One aspect that I do want to mention, as

 

      we think about this, the patient has to be foremost

 

      in our minds, what are the unmet needs, and as we

 

      sort of develop this, I think clearly, the patient

 

      needs have to be kept in mind as we move forward,

 

                                                                19

 

      because there are many diseases, many aspects where

 

      we don't have effective drugs, and so forth, so we

 

      shouldn't forget that aspect.

 

                DR. KIBBE:  Marvin, are you ready now?

 

                DR. MEYER:  Yes.  I was just thinking of a

 

      simple example where the Agency I think played a

 

      major role and really expedited drug approval, and

 

      that was back when we were battling over assay

 

      method validation.

 

                The hypothesis was if we had a better way

 

      of validating assays or a uniform way of validating

 

      assays, things would get approved without recycling

 

      and redoing, and, in fact, FDA then, and APS and

 

      others, convened several workshops, had white

 

      papers, ultimately put out a guidance, and I

 

      suspect that hypothesis has been tested, that there

 

      are much fewer problems in the local methodology,

 

      so I think that is a good model, and you alluded to

 

      that.

 

                DR. KIBBE:  Anybody else would like to

 

      make a comment?

 

                DR. SINGPURWALLA:  Yes.  I repeatedly hear

 

                                                                20

 

      from individuals like yourself asking industry to

 

      share more information with you.  What is the

 

      incentive to the industry to do so, because there

 

      is a penalty to do so?  Recently, those of us who

 

      read the Washington Post can see the number of

 

      pages devoted to the Merck and also to Pfizer

 

      having a similar drug going to be tested, and

 

      things like that.

 

                So, unless the legal pressures that are on

 

      industry are defused or removed, industry is going

 

      to be foolish to share all the information with

 

      you.  I wouldn't. It's like me going to the IRS and

 

      saying look, this is how I have cheated, catch me.

 

      It doesn't make sense, does it?

 

                DR. HUSSAIN:  No, I think it is not in the

 

      context of sort of cheating, and so forth.  This is

 

      in the context of how much we know and how much we

 

      don't know, to start filling the gaps where the

 

      knowledge exists.  Clearly, that is the aspect, and

 

      it is complicated by the fact that the way it gets

 

      entrenched into the legal and political scenarios,

 

      those are significance challenges, no doubt about

 

                                                                21

 

      that.

 

                DR. SINGPURWALLA:  What is the industry's

 

      response to this?

 

                DR. MIGLIACCIO:  Well, it is complicated

 

      by obviously, intellectual property rights, which

 

      is the life blood of a commercial business, but it

 

      is also complicated by--you just used the word

 

      "trust."

 

                I will give an anecdote here.  I went to

 

      an internal FDA meeting to provide training, and

 

      during that training, provided knowledge about

 

      products, which normally, would not have been made

 

      available.

 

                The reaction was the traditional

 

      predictable reaction, not the forward thinking

 

      reaction, by certain elements of the audience.  So,

 

      that was a risk, that was a poorly thought-through

 

      risk on my part.

 

                We have to reduce the risk associated with

 

      sharing knowledge.  That is the fundamental issue

 

      is if we share knowledge and expose ourselves to

 

      compliance action where that knowledge is

 

                                                                22

 

      essentially reflecting what is the scientific

 

      truth, and we can now measure that, we can now see

 

      that where we couldn't before, and if you divulge

 

      that knowledge and risk compliance action versus

 

      scientific discussion, then, the knowledge will not

 

      be transferred.

 

                DR. KIBBE:  Ajaz, anything?

 

                DR. HUSSAIN:  No.

 

                DR. KIBBE:  Anybody else?  Let me just put

 

      three things on the table and perhaps you can think

 

      about them as how they respond to the questions we

 

      were left with yesterday.

 

                One is that I think I heard from around

 

      the room that the Agency has a limited resource

 

      base, and it truly should focus on those aspects of

 

      the critical path that only the Agency can do, that

 

      no one else has the wherewithal or the capability

 

      or the information to do that.

 

                Secondly, that we try to get others who

 

      are even more capable of responding to certain

 

      aspects of the critical path to take on that

 

      burden.  I am thinking primarily of industry and

 

                                                                23

 

      perhaps industry/academia together looking at those

 

      aspects of it.

 

                But the third thing that I think that the

 

      Agency and both the industry will have to look

 

      forward to is that the rate of technological

 

      advance is such that 10 years from now, the

 

      questions that you are trying to answer now will be

 

      ancient history, and the questions that you are

 

      running into are going to be dramatic and clearly

 

      different, and I really look forward to a paradigm

 

      shift in the way we approach therapy, and I would

 

      recommend to the industry that they change their

 

      name from drug companies to companies that provide

 

      therapeutic agents and processes, because they

 

      could be caught up in the same system that the

 

      railroads did.  They were railroad companies, and

 

      not transportation companies.

 

                I don't know how the Agency can respond

 

      effectively without having some type of internal

 

      committee that is constantly looking at four or

 

      five years out and the technology that they are

 

      going to have to deal with then.

 

                                                                24

 

                So, that is where I think the critical

 

      path kind of initiative ought to be looking.

 

                DR. DeLUCA:  Let me just comment.  I made

 

      some notes here from yesterday, and I think that

 

      this is based on collaboration, I think I am going

 

      to really focus on that, and as Jerry mentioned, I

 

      think trust.  Certainly, trust is essential in

 

      collaboration, and as we get into talking about the

 

      science-based approach here and research, research

 

      is a search for the truth.

 

                I would like also to commend the Agency in

 

      their research efforts.  I mean yesterday was, I

 

      think, I would say overwhelming to learn the type

 

      of research that is going on and the collaboration

 

      with NIH, so I really have to commend the Agency

 

      for this.

 

                I would like to also talk about the

 

      presentation by Monsoor Khan where he talked about

 

      critical path research and some of their efforts,

 

      and I think Gerry Migliaccio had responded that

 

      industry takes this approach.

 

                I have to say that that is true from my

 

                                                                25

 

      experience to an extent.  I know, I am involved in

 

      the novel drug delivery area and the research in

 

      that area, and working with a company that was

 

      scaling up or transferring some technology, that

 

      that approach was taken, the critical path approach

 

      was taken, and worked with them, and they did solve

 

      the problem at hand, but there were other things

 

      that still needed to be done to define some of the

 

      process variables, that once the problem was

 

      solved, they went on, they didn't want to go any

 

      further with that.

 

                So, I think there is a limit to where they

 

      go and I think this is where collaboration is

 

      important, and I think there is a need to continue

 

      on and to search those things out, and probably the

 

      place for that is in academe.

 

                I don't know if it was Jerry Collins, when

 

      he talked about the science-based approach to

 

      critical path issues and the research, that it is

 

      probably essential that the research that is going

 

      on, that it is going to be hypothesis driven.  I

 

      think this is something that many times the

 

                                                                26

 

      research that takes place, and if it is in an

 

      industrial setting, may lack the hypothesis driven

 

      type of research, and that probably I think is

 

      important.

 

                I guess my feeling is in hearing all the

 

      things, and I think what Art had said, FDA can't

 

      really overreach, I mean there is a limit

 

      resourcewise, but I think more importantly is the

 

      idea that they can't do it alone, so I think that

 

      collaboration is important.

 

                The FDA has been collaborating more with

 

      NIH, and I think translational research issues,

 

      taking the drug product development, that portion

 

      of it along, but I think there is a gap there with

 

      the critical path, in the formulation and taking it

 

      and the manufacturing science, and I guess I think

 

      that the collaboration has to be there between

 

      academe, industry, and FDA, and FDA could really

 

      set the stage for this.  I think there is a very

 

      important role.

 

                Just to bring out my experience with the

 

      journal, the APS on-line Pharmaceutical Science

 

                                                                27

 

      Technology Journal, that the submissions, we get

 

      about 50-50 from abroad and the United States, but

 

      about 90 percent of the submissions--now, this is

 

      in the Pharmaceutical Technology Journal, and you

 

      would really think that you would get more from

 

      industry--but about 90 percent of the submissions

 

      come from academe.

 

                I have to acknowledge that probably in

 

      about 40 percent of those, there is a collaboration

 

      between academe and industry, so that there is a

 

      tie-in and that it is all those being submitted

 

      from the academic institution, the industry is

 

      involved in it.

 

                But I think that this kind of sends a

 

      message, and I have tried to encourage more, more

 

      submissions from industry, and there is

 

      intellectual property situations involved in that,

 

      but I think there is a need.

 

                I know, being in academe and graduating

 

      Ph.D.'s, the majority of them will go into the

 

      industry, few of them publish after they are in

 

      industry.  Before they left, they had eight or nine

 

                                                                28

 

      publications, and then they went in and stopped

 

      publishing, so, I am not sure that is a good thing.

 

                What I wanted to emphasize here is that

 

      there is an essential need for collaboration.  I

 

      think the whole science-based approach to the

 

      regulatory arena is great, and I have to commend

 

      the Agency in this, but they just can't do it

 

      alone, and I think there is an essential need that

 

      this collaboration occur.

 

                It may be that with that type of

 

      collaboration, you know, for a long time in

 

      academe, we have talked about the NIH and trying to

 

      get them involved with supporting drug product

 

      research and development to little avail, so maybe

 

      now is the time.

 

                Certainly, I think it is essential that

 

      this type of approach be taken in the manufacturing

 

      sciences, because it certainly will benefit, I

 

      think, our society, so I think it is in the

 

      interests of the country, so that hopefully, the

 

      NIH will look a little bit more favorably on

 

      supporting this type of research.  I think the

 

                                                                29

 

      collaboration between FDA and NIH may help in doing

 

      just this.

 

                DR. KIBBE:  Thank you.

 

                Ken, go ahead.

 

                DR. MORRIS:  Thanks, Art.  Welcome to your

 

      last day.

 

                A couple of things I wanted to say first

 

      to Nozer's point.  In the face of the data that I

 

      think Merck generated or was generated and then

 

      shown to Merck, I don't think they would need the

 

      Agency to tell them that they needed to pull the

 

      drug or to modify it.  They are really very

 

      responsible about that sort of thing.  I understand

 

      your point.

 

                DR. SINGPURWALLA:  The lawyers made them

 

      do it.

 

                DR. MORRIS:  Well, the lawyers made them

 

      do it, but the drug companies in general, the

 

      innovators of generics, when they see problems like

 

      that, are still I think honor bound and have

 

      historically done a good job of monitoring

 

      themselves with respect to public health when there

 

                                                                30

 

      is a clear and present public health injury issue.

 

                But beyond that, let me just comment, if I

 

      can, I have just five points here, relatively

 

      short.

 

                The first is, is that the unique

 

      opportunity afforded by the FDA massive database, I

 

      think is absolutely invaluable and needs to be

 

      exploited to the maximum.  I mean that, in my mind,

 

      is perhaps the number one initiative in terms of

 

      getting down the critical pathway with all due

 

      deference to proprietary data, of course, as we saw

 

      yesterday.

 

                There are some issues I think, for

 

      instance, tox, where the database would probably

 

      not be nearly as good as even the sampling of the

 

      Big Pharma companies' databases on tox, because you

 

      don't have the tox information on compounds that

 

      never made it to filing, so they may actually have

 

      a bigger database there, which would really help in

 

      the really interesting work we saw yesterday on

 

      modeling.

 

                The second point is it look from a

 

                                                                31

 

      nonbiological and obviously blue collar tablet

 

      smasher, that there is a fairly large disparity

 

      between the amount of internal biological research

 

      versus product development research.  I am not

 

      really in a position to judge what the priorities

 

      in the biologicals should be.  It is all obviously,

 

      very high-caliber research.

 

                I am not in any way commenting on that,

 

      nor am I capable of it, but I think it does point

 

      out the fact that there are developmental research

 

      agendas that probably would be better handled in

 

      part at least, or at least administered through the

 

      Agency, that aren't being, and we can talk about

 

      specifics, and have with John and you and others,

 

      of course.

 

                But I think that points out an opportunity

 

      if you were on the critical path, and that is the

 

      prioritization that I was asking about yesterday,

 

      is that given the breadth of projects and the

 

      dearth of resources, I think the prioritization,

 

      particularly the internal research projects,

 

      becomes your biggest challenge and one that I think

 

                                                                32

 

      could be helped by the committee, and I think has

 

      been in part, hopefully, in these days.

 

                It also points out, to reinforce Pat's

 

      point, and this is a little bit self-serving, but

 

      the amount of research that doesn't or is not as

 

      logically done within the Agency, needs to find

 

      federal support in terms of public health

 

      initiatives, as well as the obvious advance of just

 

      basic science.

 

                To address a question that Gerry had

 

      raised with respect to the possible putative

 

      consequences of sharing information, there is a

 

      mechanism that we have been sort of developing,

 

      which is to, through blinded intermediates, to be

 

      able to discuss general topics without filtering.

 

                I am not talking about filtering data or

 

      hiding data, but to bring data to light to the

 

      Agency in a blinded manner to say, you know, is

 

      this the sort of data that would be useful, or is

 

      this the sort of data that would give you cause to

 

      think that there was no particular reason to review

 

      it, and it would just be a waste of the Agency's

 

                                                                33

 

      time.

 

                So, I think there are mechanisms to do

 

      that.  They are not formal mechanisms, but through

 

      consultants and whatnot, I think you have already

 

      got that as an opportunity, so you can't be too

 

      specific, of course, because once you are too

 

      specific, then, you have already revealed what it

 

      is you are asking about.

 

                Finally, with a question of metrics, I

 

      think the suggestions you made yesterday, the

 

      multiple review cycles I think is a great metric.

 

      That was what the Manufacturing Subcommittee, at

 

      Judy's last meeting, we talked about the idea that

 

      in the new or the desired state, instead of having

 

      minimal data that the reviewers have to try to

 

      piece together into some sort of Frankenstein

 

      rationale, if you get the rationale in a piece from

 

      the companies with summarized supporting data to

 

      make it a compelling argument, then, the reviewers

 

      just have to assess the sufficiency of the

 

      rationale as opposed to trying to piece together

 

      one on their own.

 

                                                                34

 

                So, I think the review cycles are an

 

      excellent metric.  The time to approval, of course,

 

      is a low hanging fruit there in terms of a metric

 

      although it is not independent, and for generics,

 

      of course, that is compounded by the workload

 

      itself.

 

                Maybe you could normalize it by

 

      normalizing the time to approval to the number of

 

      pre-filing and pre-approval meetings, the off-line

 

      meetings that John talked about yesterday, John

 

      Simmons talked about yesterday.

 

                The other one--and I don't know if we have

 

      talked about this before--is to track FDA personnel

 

      turnover.  I think it is not a bad metric to look

 

      at retention of the FDA reviewers themselves.  I

 

      mean it is a very high-pressure job, it is not all

 

      that celebrated a position, but obviously of key

 

      importance.

 

                I think that does two things.  One is it

 

      gives us a metric of how effectively the program

 

      works, and the other is that it gives an internal

 

      metric for the personnel management, so you don't

 

                                                                35

 

      burn out your best and brightest.

 

                DR. HUSSAIN:  Ken, the whole aspect of the

 

      critical path was in a sense that review cycles

 

      have really come down, so that review cycle is not

 

      the rate-limiting step in the critical path.  So,

 

      there are different metrics for that purpose.

 

                DR. KIBBE:  Marvin, do you have something

 

      else?

 

                DR. MEYER:  A quick comment.  Helen was

 

      saying last night that on the ANDA side, that the

 

      generics are now, or shortly going to be, required

 

      to submit all studies they did, not just the 1 out

 

      of 12, the test.

 

                Maybe, and this is terribly naive because

 

      I don't know all of the complications, but maybe

 

      there is some way down the line of having the NDAs

 

      be accompanied by a synopsis, at least, of what

 

      they tried and what failed, a one-pager perhaps.

 

                We tried doing virus filtration this way,

 

      and it failed because, we think it because, and

 

      this might be attached with the NDA, but reviewed

 

      independent of the NDA. There might be a group at

 

                                                                36

 

      FDA that evaluates failures, if you will.  So, some

 

      way of getting the data to the Agency that wouldn't

 

      impact on the NDA and yet would provide the Agency

 

      with I think some valuable information.

 

                DR. KIBBE:  I am concerned that as much as

 

      we in academia value getting all the information,

 

      industry values having information that their

 

      competitors don't have, and if they have a lot of

 

      failures they corrected, and they know what

 

      mistakes not to make, they generally think they

 

      have an edge on doing it right, and they are not

 

      really excited about turning that over to someone

 

      else, let them make their own mistakes and figure

 

      it out.

 

                I think the time that we will actually be

 

      able to share all the information about all the

 

      drugs that have ever been approved is when Glaxo

 

      finishes buying everybody or Pfizer has merged with

 

      whoever is left, and there now is International

 

      Therapy Development Company.

 

                DR. KIBBE:  Ajaz, anything to wrap up

 

      with?  Okay.

 

                                                                37

 

                DR. SINGPURWALLA:  Mr. Chairman, I have a

 

      few thoughts.  Ajaz, back.

 

                [Laughter.]

 

                DR. KIBBE:  It's okay, Ajaz, you can

 

      escape if you would like.

 

                DR. SINGPURWALLA:  Ajaz, you asked three

 

      questions here.  To be quite honest with you,

 

      yesterday, I couldn't focus on these because I

 

      couldn't get my mind straight as to what we are up

 

      to and what is happening.

 

                But subsequently, I think I can answer

 

      some of your questions very directly.

 

                I looked at TR Critical Path Initiative

 

      Challenges document, and to be quite honest with

 

      you, I think you are on the right track, and I

 

      think you are thinking along the proper lines.

 

                Two, three things come to my mind.  Your

 

      mention or at least the mention of design of

 

      experiments that was discussed is one of the right

 

      ways to go about things.

 

                You also mentioned the use of bayesian

 

      ideas. That  is the best way to reduce time cycles

 

                                                                38

 

      because you are taking advantage of all other

 

      sources of information, but you don't want to use

 

      that only for clinical trials, but you want to use

 

      it throughout the entire process.  Again, you have

 

      highlighted it, so I think again you are on the

 

      right track.

 

                The one thing is you cited examples from

 

      manufacturing.  That is fine, but I seriously

 

      consider you also look at the area of weapons

 

      development.  They face problems very similar to

 

      yours and you may want to see what they are doing

 

      and how they are developing their particular

 

      processes, and the weapon development process has

 

      much in parallel.  The two communities are very

 

      alien to each other, but I urge you to look into

 

      what they are doing, and I think I can say that you

 

      are on the right track.  You are focusing on the

 

      issues that I would focus about, that is all.  I

 

      wanted to reaffirm it.

 

                DR. HUSSAIN:  Thank you.

 

                DR. KIBBE:  Paul.

 

                DR. FACKLER:  Let me just offer a couple

 

                                                                39

 

      of thoughts to those questions, you know, are you

 

      on the right track.  Of course, I am speaking for

 

      the generic industry, but it is difficult to give

 

      people help when they haven't asked for any, and I

 

      can't speak for PhRMA, and I don't know if PhRMA

 

      has come to the Agency and said, help, we can't

 

      develop new drugs.

 

                So, I think you face a very difficult

 

      challenge trying to assist a process that maybe the

 

      people actually doing it don't feel is broken.  The

 

      economics of drug development in 2004 is

 

      significantly different than it was in 1994.

 

                You know, if you have a company selling

 

      $50 billion in drugs a year, and they want to grow

 

      by, say, 5 percent, which isn't acceptable by any

 

      means, they need to get an additional $2 1/2

 

      billion in revenue out of the new drugs that they

 

      are developing.

 

                So, you know, a product that has some

 

      marginal value, say, 50- or $100 million that would

 

      benefit society probably just gets put in an

 

      envelope somewhere, and not brought out.  It is a

 

                                                                40

 

      problem with the situation in industry, but I am

 

      not sure FDA is going to be able to do anything to

 

      assist that.

 

                Let me speak to the generics because there

 

      was a presentation yesterday, and speaking for the

 

      generic industry, we have communicated with FDA

 

      where we think we need help.  We have asked about

 

      topical products, we have asked about inhaled

 

      products, biologics, of course, are an issue, and

 

      time to approval is a real issue for us.

 

                So, the question was are you on the right

 

      track, and at least from the generic perspective,

 

      the answer is yes.  I think you are trying to

 

      overcome the hurdles that we face, that would

 

      assist us in bringing products to the market

 

      earlier.

 

                I know it is not really the main thrust of

 

      the Critical Path Initiative, but for our portion

 

      of it, the answer is yes.

 

                DR. KIBBE:  Thank you, Paul.

 

                DR. HUSSAIN:  I think just the point

 

      generics are equally important for us, so they are

 

                                                                41

 

      part of the critical path from an OPS perspective.

 

                DR. KIBBE:  Gary.

 

                MR. BUEHLER:  Well, we have had a number

 

      of mentions of our workload.  It is significant.

 

      We did receive 563 applications I believe the last

 

      fiscal year, 449 the year before, and 361 the year

 

      before, so we are increasing by about 100 a year.

 

                It is a bit scary, but we are dealing with

 

      it, and we are communicating with the industry

 

      significantly on what we can do to make their

 

      applications better and to make our responses to

 

      them more predictable, so that they know what we

 

      want.

 

                As part of the critical path, and we are

 

      trying to work in providing the information that

 

      the industry needs to develop their products, and

 

      this is through the dissolution methods and the

 

      bioequivalence methods that we get tons of letters.

 

                We got over 1,000 correspondence last

 

      year, over half of them requesting what is the

 

      bioequivalence method for a particular drug, what

 

      is the dissolution method for a particular drug, so

 

                                                                42

 

      we can begin to develop our products.

 

                We are trying to get that up as a

 

      web-based program, so that they can actually access

 

      these methods.  We have people working full time in

 

      our office to research this information, so we can

 

      make it available to the firm.

 

                Now, this isn't revolutionary stuff.  This

 

      is stuff that we have always provided them.  We

 

      just want to provide it to them faster.  We want to

 

      make it easier for them to access this information,

 

      and we don't want to get as many letters.  The

 

      letters that we get obviously take up our resource

 

      time, and we want those resources to be put toward

 

      application review.

 

                We hope to be able to get these up soon.

 

      I know I promised them I think six months ago to

 

      the industry.  Things are never as easy as you

 

      would like them to be in the Agency.  A lot of

 

      people have to sign off and make sure that we are

 

      not giving away the farm, and we don't want to give

 

      away the farm, but we do want to give away

 

      information that is needed by the generic industry.

 

                                                                43

 

                The generic industry is a very viable,

 

      very robust industry right now.  A lot of new

 

      players are getting into it, a lot of people want

 

      to put applications in as evidenced by our

 

      workload.  We welcome that workload, we are glad.

 

      This country needs generic products.  A lot of

 

      people out there can't afford prescription drugs

 

      out there.

 

                So, we welcome the work and we welcome the

 

      challenge.

 

                DR. KIBBE:  Anybody else?  Okay.

 

                We have an opportunity now to hear from an

 

      absolute genius.  They asked me to give a talk on

 

      visionary overview, and I will get up there, then,

 

      I will pontificate for half an hour, and I hope you

 

      all enjoy it.

 

               Science in Regulation - Visionary Overview

 

                DR. KIBBE:  I need a soapbox.  I have six

 

      slides.  This is to reduce some of the slide

 

      overload that we are suffering from.  You all have

 

      copies of these slides.  You can tell that the

 

      slides are really informative because they are

 

                                                                44

 

      filled with words.  I look at slides and I say,

 

      hey, there are 22 slides and each one has 180

 

      words, how am I going to get through it, so I put

 

      up a couple of simple slides.

 

                First, the title was given to me by the

 

      Agency.  I looked at it and I said Visionary

 

      Overview, I guess they think I am a visionary, why

 

      would they think that.  So, I thought long and hard

 

      about why they think I am a visionary, and I

 

      realized it was because I live in Pennsylvania,

 

      which is the home of the world's most well known

 

      and renown visionary, the seer or all seers, the

 

      procrastinator for all good things, Punxsutawney

 

      Phil, who comes out and tells you whether you are

 

      going to have winter for another six weeks or not.

 

                I also would like to make a disclaimer, we

 

      do lots of disclaimers.  All the ideas that I

 

      express today are strictly my ideas, and I would

 

      not saddle anyone in the scientific community and

 

      industry over the Agency with any of these

 

      cockamamie ideas.  So, they are all mine and

 

      hopefully, they will stimulate your thinking

 

                                                                45

 

      without putting you completely to sleep.

 

                So, what has the FDA and we been doing for

 

      the last few years?  I actually went out and got a

 

      copy of the agenda for the first meeting I was at,

 

      and there wasn't PAT mentioned in the agenda, but

 

      when you looked through the agenda, you saw the

 

      beginnings of what was I think a wonderful three-

 

      or four-year push in an area that can significantly

 

      impact industry's bottom line, and hopefully, the

 

      industry will be in a mood of generosity and have

 

      that bottom line, some of those savings reflected

 

      in the cost of goods produced.

 

                The effort I think was an opportunity for

 

      me to view the way that scientists from industry,

 

      both the generic and innovator companies,

 

      scientists within the FDA, and scientists from

 

      academia, and those consultants who serve all of

 

      us, could get together, look at a problem, develop

 

      a reasonable approach to it, something that would

 

      work in the community that we work in, and really

 

      come up with something worthwhile.

 

                I could go on about the successes we have

 

                                                                46

 

      had, but they don't make great news, and the news

 

      media always wants failures and disasters to report

 

      on, and so I will move directly into those.

 

                First, is it the Agency's role to apply

 

      science to regulation?  Of course, we all agree it

 

      is.  The application of the scientific method to

 

      goalpost generation for the industry is extremely

 

      important, and I am going to try to look at what we

 

      have done and where we are going, and perhaps make

 

      some projections out.

 

                If we are going to regulate a

 

      science-based industry with science, then, we need

 

      to use a scientific approach to where we are going.

 

                We all are familiar with linear

 

      regression, and we know that there is a certain

 

      amount of error associated with it, but in order to

 

      project beyond the data that we already have, we

 

      have to have a significant amount of data going

 

      backwards to draw a line through, so that as we go

 

      out in the future, we get closer to the truth.

 

                We know that the further out in the future

 

      we can project, the less reliable the answer is,

 

                                                                47

 

      but we do it anyhow, and I am going to do that.

 

                So, where have we been in terms of

 

      regulating the quality of drug products and

 

      therapies in the United States? Of course, we start

 

      in 1817 with Dr. Spalding, and he decided that we

 

      ought to get the physicians together and say why

 

      can't we have quality products to give to our

 

      patients, let's set up some standards, and the USP

 

      was formed.

 

                So, we started the regulation of the

 

      quality of how we treat our patients by getting the

 

      health care providers who treated patients together

 

      to decide what quality was and how to arrive at it.

 

                After the Civil War, the pharmacists got

 

      together and decided that while the USP had

 

      standards for individual ingredients, it really

 

      didn't have standards for how to mix them together

 

      and make them useful, so they decided to publish

 

      the National Formulary, and I was instrumental in

 

      the first edition, and I brought my copy with me.

 

                This is the sum total of how to make

 

      pharmaceuticals in 1888, and compare it with what

 

                                                                48

 

      we know today and how many shelves it takes up, and

 

      how controversial each little, tiny issue is.  Of

 

      course, we also know that you have to learn Latin

 

      to use this, so it's dead along with the dead

 

      language that it is written in.

 

                At the same time, the industry actually

 

      regulated itself.  There was a comment made here a

 

      little while ago, which said that lawyers make them

 

      do things.  I would argue that in the current

 

      litigious society, companies act slower to remove

 

      drugs from the market when they have worrisome data

 

      than they would if there wasn't a litigious

 

      society.

 

                I think they worry more about what it

 

      means to their future class action suits to

 

      actually admit that there is a problem until they

 

      have all their lawyers lined up, so they know how

 

      to defend themselves, and if they weren't worried

 

      about the fact that the American public has an

 

      exaggerated misconception of what drugs do and

 

      work, they would act quicker.

 

                I think the American public in general

 

                                                                49

 

      expects drugs to be safe and effective, and they

 

      don't recognize that drugs can be safe and

 

      effective if used correctly, but in the wrong way,

 

      are dangerous and shouldn't be used, and they don't

 

      get that.  They just don't get it.

 

                I put E.R. Squibb down because I know a

 

      little bit about E.R. Squibb as an example of the

 

      leadership that the industry had back in the 19th

 

      century.  Dr. Squibb, a physician, wanted a higher

 

      quality ether for anesthesia.  This was an

 

      extremely important drug in those days, and so he

 

      founded a company for the express purposes of

 

      making sure he had high quality ether.

 

                He built it in Brooklyn, and then his

 

      company started making other things and then he

 

      noticed that there were other companies that were

 

      copying his products, calling them the same thing

 

      and putting them out there less expensively, and he

 

      said the public might be at risk if they aren't

 

      made correctly.

 

                So, he did something unique which I don't

 

      think any of the companies would do today.  He got

 

                                                                50

 

      all his formulas together, how he made everything,

 

      and he published them in the Journal of the

 

      American Pharmaceutical Association with the

 

      proviso that if anybody wanted to make a product

 

      that E.R. Squibb sold, they should make it the way

 

      we make it, so it would be of the same quality, so

 

      at least the public would have a good quality

 

      product, and if they could make it less expensively

 

      than we could, good luck to them.

 

                Well, I wonder how many companies are

 

      ready to jump into that game.  At that same time,

 

      of course, Eli Lilly was producing well over 100

 

      generic products.  It was the largest generic

 

      manufacturer in the United States.  It produced

 

      everything that could be made that was listed in

 

      the USP or NF, extracts, and what have you.  It was

 

      an interesting time.

 

                Now we get into government regulation.

 

      Now, why did the government get into regulation?

 

      Well, it bought quinine that wasn't quinine and it

 

      got upset.  So, in 1848, with the troops attacking

 

      Mexico City, their quinine didn't work like it was

 

                                                                51

 

      supposed to, they said what's in here, it wasn't

 

      quinine, it was something else, I don't know what

 

      it was.

 

                They said that's terrible, terrible,

 

      terrible, and so we needed to find a way to make

 

      sure that when something was labeled quinine, it

 

      really was indeed quinine.  That was the first shot

 

      out of the cannon.

 

                We finally had the Food and Drug Act of

 

      1906, which really just said that if you are going

 

      to sell something and call it a drug, and name it,

 

      it ought to be what you call it.  Right about that

 

      time we got into the concept of misbranding, which

 

      was putting something in something and calling it

 

      something that it wasn't, and that is basically

 

      what misbranding is.

 

                We have a lot of meetings for misbranding

 

      now, but the bottom line is that it is not what it

 

      is supposed to have been.

 

                The Agency wasn't really founded then, but

 

      the government said that if we wanted to take

 

      action against the company that misbranded a drug,

 

                                                                52

 

      that it was incumbent upon the government to prove

 

      that the drug was indeed not what it said it was,

 

      and that there was intent to defraud.  If you do

 

      that to the government, we can't enforce any

 

      quality on anybody, because we don't have any

 

      information to use for it.

 

                But I want you to remember that concept

 

      that came about in the early 1900s, because when we

 

      get to the end of the 1900s, we have another law

 

      that brought us right back to that place.

 

                So, 1938, we killed a bunch of kids in the

 

      New York City area with antifreeze as a sweetening

 

      agent in a sulfa drug preparation.  That was the

 

      end of a company's reputation, and well it should

 

      have been, and everybody was in an uproar, so we

 

      now have a new regulation.  You will notice the

 

      trend here - disaster, new regulation, disaster,

 

      new regulation.  It's kind of a recurring theme.

 

                So, we know said, okay, it has to be what

 

      it says it is, it has to contain what it says it

 

      contains, and it has to be safe, but it doesn't

 

      have to work.

 

                                                                53

 

                Homeopathic remedies are exactly that.

 

      They are 1 to 100 dilutions of something done 1,000

 

      times.  You end up with a bottle of water, which

 

      they claim contains the essence of the power of

 

      whatever drug was in the first bottle of 1,000

 

      dilutions before.  All right.  So, we can claim it

 

      works, and it contains a diluted, diluted, diluted,

 

      fine, that is what it really contains.  You can't

 

      find a molecule because you have diluted more than

 

      Avargordo's number, so we have products on the

 

      market.

 

                By the way, the Food, Drug, and Cosmetic

 

      Act says specifically that drugs are things that

 

      are contained in the homeopathic pharmacopeia,

 

      which means that they are precluded from acting

 

      against products that are in the homeopathic

 

      pharmacopeia even though we know they don't work.

 

                We are still working with things that are

 

      just safe, but at least they are branded right, you

 

      know.  Nowadays we have people who claim that water

 

      solves medical conditions.  What the heck, you

 

      know, 1938, that would have worked, put a label on

 

                                                                54

 

      water, say, if you will bottle water and pay for it

 

      at a rate higher than you pay for gasoline, then,

 

      it is better for you than the water you get for

 

      free out of the tap, and you will do better.

 

                Well, I don't know, I wonder about things

 

      like that.  I have a problem my students always

 

      complain about.  I have one of those minds that

 

      kind of wanders, and so I do that.

 

                Let's get back to misspelled words and

 

      regulation. So, in 1951, two pharmacists got

 

      together, a guy named Carl Durham and a guy named

 

      Hubert Horatio Humphrey--I love his name.  They

 

      were pharmacists.  One was in Congress and one was

 

      in the Senate.  Hubert came from Minnesota.  He

 

      ultimately became vice president, ran for

 

      president, didn't make it.

 

                I often wonder what would happen if the

 

      president of the United States was really a

 

      physician or a pharmacist, a health care worker,

 

      what difference that would make in their approach

 

      to the health care problems.

 

                So, they got together and they said, you

 

                                                                55

 

      know, there is a lot of drugs out there that are

 

      pretty dangerous, that the average person really

 

      can't understand, and maybe we ought to have

 

      somebody help them figure out what to take, so they

 

      established two criteria, prescription drugs and

 

      over-the-counter drugs.  We still, by the way,

 

      don't have to have them work.  You know, God

 

      forbid, they actually should work.

 

                We are a unique country among the

 

      developed nations of the world.  We only have two

 

      categories of drugs. Most of them have many more

 

      categories of different levels, and, in fact, I

 

      like the Australian system.  They are listed in the

 

      group of things they call poisons, so we clearly

 

      know where they belong, right?  They are the poison

 

      list.

 

                In 1962, we finally got around to hoping

 

      that we could figure out that the drugs were both

 

      safe and effective, so in 1962, we said, okay, new

 

      drugs have to be safe and effective.  The Agency

 

      was kind of curious.  It said, but you can't tell

 

      people that this is an approved drug, because that

 

                                                                56

 

      gives you a marketing advantage over the drugs that

 

      haven't been approved by us, and then we don't know

 

      are effective.  Hmm, that's interesting.

 

                Then, Congress, in its infinite wisdom,

 

      jumped right in there with DSHEA, and DSHEA says

 

      that if you aren't really a drug, but kind of imply

 

      that you are a drug, then, you can go back to the

 

      1906 regulation which says that it only has to be

 

      what it says it is, and it doesn't have to be

 

      proven to be safe or effective, and if there is any

 

      problems with it, the Agency has to compile the

 

      data before they can make you take it off the

 

      market.  I just love that, you know, retrograde

 

      regulation, I just wonder about the wisdom of that.

 

      I am sure it has to do with the need that the

 

      public has for unsubstantiated claimed herbal

 

      remedies.

 

                All right.  Here is where we really get to

 

      where the rubber meets the road, and that is the

 

      cost of drugs.  I grew up in a pharmacy family.  My

 

      father was a pharmacist, my uncle was a pharmacist,

 

      I became a pharmacist because I didn't know

 

                                                                57

 

      anything else.

 

                I grew up in a drugstore, and when I was

 

      at a young age, I worked in my father's drugstore

 

      as a soda jerk. Some people think that I have never

 

      gotten over the second half of that.

 

                But in those days, the average cost of

 

      drugs that my father filled--he has a wonderful

 

      ledger, handwritten in ink pen where he wrote down

 

      the name of the patient, the prescription, the

 

      physician, and then the cost--and if you look at

 

      it, you will find that the average charge to his

 

      patient was $1.75.

 

                I asked him one day, being a nosy

 

      teenager, how do we make money to live on at the

 

      store here, and he says, "Well, I charge $1.75, but

 

      it costs me about 25 cents of goods."  So, I said I

 

      thought that was pretty good.

 

                Of course, nowadays, the average charge of

 

      a prescription can be in the $50 or $60 range, and

 

      the pharmacy gets $3.50.  There has been a shift

 

      here somewhere.

 

                At that time, Tetracycline came out.  It

 

                                                                58

 

      was about 50 cents a capsule.  The price of

 

      Tetracycline has gone down dramatically, but we

 

      keep bringing out new drugs, and I think each time

 

      we bring out a new drug, we say what was the price

 

      that we charged for the last new drug, and we

 

      multiply by 1.5.

 

                You also understand that there is

 

      absolutely no relationship between the charge for

 

      the drug and the cost of actually manufacturing it,

 

      and that they factor in all of the other costs to

 

      maintain the corporate entity that creates new

 

      drugs.  So, they need to have this huge inflow of

 

      money in order to float all of the research and the

 

      marketing, and all the other efforts that go on,

 

      and so that there is some disconnect.

 

                Waxman and Hatch got together.  We

 

      recognized back in the 1980s that the cost of

 

      health care was going up quickly.  Uwe Reinhardt

 

      has a wonderful graph that he puts up, a Princeton

 

      economist, that shows the gross national product

 

      and its rate of increase and the cost of health

 

      care and its rate of increase, and then he predicts

 

                                                                59

 

      some date in the future where the two will meet.

 

                Then, he has a cartoon where he has two

 

      physicians lying in beds in the hospital together,

 

      prescribing for each other, and he said that is

 

      going to be the entire productivity of the United

 

      States is going to be this.

 

                So, we know that there is a disaster in

 

      the future and what are we going to do about it,

 

      and we have a culture in the United States where we

 

      don't regulate the price of drugs.  We are again

 

      unique.  Very few developed countries have that

 

      compunction.  So, we try to regulate it through

 

      competition.

 

                So, the Waxman-Hatch Act or the

 

      Hatch-Waxman Act, depending on whether you are a

 

      Republican or a Democrat, came into being, and it

 

      was a compromise that was supposed to benefit the

 

      innovator companies by ensuring them a reasonable

 

      patent extension or exclusivity time frame in order

 

      to recoup the investment to bring the new drug to

 

      the market, and established rules and regulations

 

      for the development of generic drugs.

 

                                                                60

 

                It seems to work in some areas and we hope

 

      for the best.  However, it is not going to be the

 

      end of the issue, and if we start to make

 

      projections out in the future, we are going to have

 

      to do more than that in terms of cost, but it was

 

      the first time that the FDA was an active

 

      participant in controlling costs.

 

                I think I see that as something going

 

      forward.  We have a problem, of course, with other

 

      issues associated with cost, and, of course, here

 

      comes re-importation, and we are going to get into

 

      that in a little bit, but I don't want to beat a

 

      dead horse.

 

                My wife is Canadian and my inlaws are in

 

      Canada, and they see the U.S. news come across that

 

      says that Canadian drugs are bad for American

 

      citizens, and they say, oh, and they call their

 

      son-in-law, the expert, and they say, "What's wrong

 

      with Canadian drugs?"  Of course, i am hard pressed

 

      to say anything about it, because there is nothing

 

      wrong with Canadian drugs.  So, that makes an

 

      interesting argument.  I think we can go down that

 

                                                                61

 

      road as long as we want.

 

                I think the next level of regulation is

 

      going to be the line on top.  People are going to

 

      want information that shows that the next new drug

 

      is not only safe and effective, but better.  I

 

      don't know how long it is going to take for

 

      Congress to do that, but that is what is coming.

 

                We have a history of producing lots of

 

      drugs that might be different, but not necessarily

 

      an improvement, where are we going to go, and I

 

      think both the industry and the Agency should be

 

      prepared to think about how they would handle that

 

      situation.

 

                Remember that we are trying to regulate

 

      according to best science, and sometimes we lose

 

      track of best science.  There are some classic

 

      equations that we use that we depend upon to help

 

      us decide what is good science.  One is the

 

      Noyes-Whitney expression.  The Noyes-Whitney

 

      expression describes dissolution profile, and it

 

      was developed by these gentlemen using a very

 

      interesting standard material.  It was a fused

 

                                                                62

 

      cylinder of material.

 

                So, their apparatus and how they did it

 

      were standardized based on one solid hunk of an

 

      individual chemical in the cylindrical form, so

 

      they could accurately determine the area exposed to

 

      the fluid and therefore, from it, determine all of

 

      the equations.  Nowadays we use a standardized

 

      compressed tablet.  I would argue that the

 

      dissolution apparatus is probably less variable

 

      than an individual tablet coming off a tablet run.

 

                If you wanted to standardize an apparatus,

 

      you ought to standardize it with something which is

 

      less variable than the apparatus you are

 

      standardizing.  I wonder about that.  I guess we

 

      could ask our colleagues down the street what they

 

      think about that, but let's go back to the basic

 

      science and figure out what is going on.

 

                The other one I like to talk about

 

      occasionally is Arrhenius.  Arrhenius developed a

 

      relationship between temperature and rate of

 

      reaction that was developed for reactions that

 

      happened in dilute solutions.

 

                                                                63

 

                We apply them, same rules, too, tablets,

 

      ointments, creams, and lotions.  We put things

 

      aside for three months at elevated temperature, and

 

      we say this is going to predict what is going on in

 

      two years.  We will give you two years, just send

 

      us the real data later.  I would argue that if we

 

      went through the data that the Agency has, that we

 

      would be hard pressed to get a correlation

 

      coefficient much over 0.3 for that data.

 

                The other thing is what is the rule and

 

      regulation.  When a rule or regulation gets out

 

      there and purports to be doing something, and it

 

      doesn't, it makes you wonder.  We have a regulation

 

      that says you have to do accelerated stability at

 

      40 degrees and 75 percent relative humidity, but

 

      you can take the humidity and temperature chamber

 

      and you can put in it a tablet container that is

 

      sealed with a descant in it and do the study.

 

                That is kind of like saying let's see how

 

      fast ice cream can melt in the kitchen, but you are

 

      allowed to put it in the freezer.  I wonder, you

 

      know, I just wonder.  I am just kind of curious

 

                                                                64

 

      about those kinds of things.  You sit around in an

 

      academic office, you are a tenured full professor,

 

      what are they going to do.  You wonder about those

 

      things.

 

                I think that there is going to be a lot of

 

      international regulation.  I think that we are at

 

      the stage where the companies are truly

 

      international.  The largest provider of generic

 

      drugs in the United States is in Tel Aviv.  Most of

 

      the big developmental innovator companies are

 

      really housed everywhere.

 

                In fact, the numbers of workers at

 

      pharmaceutical plants in the world has shifted from

 

      the United States out. If that is true, then, we

 

      really have to have cooperative control on quality.

 

      I am sure that England and Germany and France want

 

      the same high quality of drugs as we do, as the

 

      Canadian Health Protection Branch insists that they

 

      do.

 

                So, we need to go in the direction of what

 

      is truly a harmonized or internationalized

 

      regulation of quality.  We need to somehow control

 

                                                                65

 

      the cost to the consumer, and if we don't find a

 

      way to do that, it will be imposed on us.

 

                One of the problems I have with all of

 

      this is that drug costs to consumer seems to make

 

      the news way more than the cost of a bed in the

 

      hospital.  Now, I will just ask you, how much does

 

      it cost to be in a hospital bed.  Does anyone know?

 

      No, but you sure know how much it costs for a

 

      bottle of Viagra--oh, excuse me.

 

                DR. SINGPURWALLA:  I don't.

 

                DR. KIBBE:  Oh, there is a man with

 

      confidence.

 

                [Laughter.]

 

                DR. KIBBE:  The reason is that most of us

 

      in the public are covered by some insurance plan

 

      that covers the cost of the hospital bed, but we

 

      aren't covered by drugs, and drugs represent 8 to

 

      10 percent of the total cost of health care in the

 

      United States, and if you look at it, it is much

 

      cheaper to give reasonably expensive drugs to

 

      patients than to put them in a hospital.  But the

 

      patients don't pay for it out of pocket.

 

                                                                66

 

                I wonder why the huge lobbying efforts of

 

      the pharmaceutical company isn't applied to getting

 

      drugs covered by Medicare and Medicaid instead of

 

      anything else. If they could ever do that, they

 

      could forget about the arguments in the newspaper

 

      about the cost of drugs.

 

                I am sure there is lots of economic issues

 

      associated with that.

 

                The last three things I have are

 

      continuous quality improvement, PAT, and

 

      federally-funded efficacy testing.  I don't know

 

      whether we are going to get the right to demand

 

      that you do an efficacy test against seven or eight

 

      of your competitors in order to get approval, but I

 

      think that the world deserves a chance to look at

 

      what is those relative efficacies in an abstract or

 

      at least impartial way.

 

                PAT has been fun for me.  I think it's a

 

      wonderful initiative, it has its own journal now,

 

      those of you who are interested in it.  It has got

 

      a forward written by--oh, my heavens--Ajaz.  It has

 

      some beautiful pictures in here.

 

                                                                67

 

                I went through it immediately and wanted

 

      to see if I knew anybody that actually was involved

 

      in PAT, and there is a whole bunch of really pretty

 

      pictures of all sorts of people that were actually

 

      on the committee with it, if anybody is interested

 

      in it.  I thought that was pretty neat.

 

                I think that there are things in the

 

      horizon that really threaten the way we do business

 

      both at the industrial level and at the regulatory

 

      level.  One of them is the development of

 

      nanotechnology and computational power.

 

                We are looking forward to a singularity in

 

      computational power, a point beyond which we cannot

 

      predict or even understand the future.  In

 

      approximately 2014 or 15, the computer on your desk

 

      will have not only digital computational power, but

 

      parallel processing, and will be able to think

 

      better than you can.  We will be able to process

 

      data, come up with new ideas, and, in fact, at some

 

      point in time, it will be the most intelligent

 

      being on the planet, and we humans will relegate

 

      ourselves to second place.

 

                                                                68

 

                When that happens, what do we do about

 

      health care?  And let's look at nanobots and what

 

      they can do.  If aging is truly a degradation of

 

      the DNA strand within people, if we can inject

 

      nanobots who know how to count DNA strands and

 

      repair them, how are we going to age?

 

                If we have the capacity to scan individual

 

      molecules and relationship in the neural net, can

 

      we then scan down a person's entire knowledge base

 

      and personality, and shift it from a carbon-based,

 

      short term, to a silicon-based, long term holding

 

      facility?

 

                How many of us would be willing at the

 

      ends of our days to become virtual us in a virtual

 

      environment?

 

                Where are we going?  Challenges to the

 

      FDA.  In our experience over the last several

 

      millennium, an ever- increasing rate of new

 

      technological development.  It was 20,000 years

 

      from the time we developed hand-held rock until we

 

      actually made a bow and arrow with a processed

 

      rock, and the rate at which we develop things now

 

                                                                69

 

      is astronomical.

 

                We need to have improved productivity in

 

      the industry, but that needs to be related to an

 

      improvement in the cost of goods sold to the

 

      American public, and the Agency needs to maintain

 

      public confidence.  It needs to not say things that

 

      are clearly difficult to defend in the public

 

      environment.  It needs to be responsive to the

 

      public needs and realistic, so that the public

 

      understands the expectations of drugs.

 

                If there was any advertising that the

 

      Agency could do, that I think would help in the

 

      long run, it is to get the American public to

 

      understand that drugs are not safe, that they can

 

      be used safely.

 

                The American public has an unrealistic

 

      expectation for their medications and an

 

      unrealistic expectation of how they should feel as

 

      they go through life, and they expect that these

 

      little pills will do it for them, and it won't, and

 

      we need to get them to stop thinking that way.

 

                We need to maintain and improve

 

                                                                70

 

      international cooperation in both regulation and

 

      harmonization, and we need to, in the final

 

      analysis, decriminalize Grandma.  When she crosses

 

      the border to pick up drugs, she needs to

 

      understand that we don't think that she is

 

      committing a heinous crime against society, that we

 

      understand that the economics are driving her to

 

      it, and we need to find a way of making it happen

 

      for her, so that she can get the drugs she needs at

 

      the price she can afford.

 

                Does anybody have any questions?

 

                [Applause.]

 

                DR. KIBBE:  Thank you, Dr. Kibbe, for that

 

      exhilarating presentation.  I am sorry, I just love

 

      those kinds of things.

 

                Now, we are going to get into some serious

 

      stuff here, because Ajaz is going to get up to the

 

      podium.

 

                DR. HUSSAIN:  Could we just take a break

 

      now and then start after the break?

 

                DR. KIBBE:  I am still fired up, you know,

 

      whatever you want to do.  You know the energy level

 

                                                                71

 

      after making a presentation.  I really want to

 

      complain about the lack of a soapbox.  I asked for

 

      a soapbox up there because I knew I was going to

 

      get on my soapbox.

 

                Ajaz wants to take a break.  Let's try to

 

      get back and get back to work at two minutes to

 

      10:00.

 

                [Recess.]

 

                DR. KIBBE:  We have comments on my talk

 

      that some members would like to make, and then I am

 

      going to be more than happy to add to my talk a few

 

      other issues, so we might have a lot of fun today.

 

                As is the tradition with this year's

 

      committee, Nozer has a comment.

 

                DR. SINGPURWALLA:  I don't have a comment,

 

      I have a question for you.  The question is what

 

      would your reaction be to the idea of nationalizing

 

      the drug industry?

 

                DR. KIBBE:  That is a wonderful question

 

      and I think the answer to it resides with our

 

      colleagues over there.  I know that if they ever

 

      did that, I would volunteer to be drug czar.  There

 

                                                                72

 

      are a couple of issues that I didn't hit on in my

 

      thing.  One of them is direct-to-consumer

 

      advertising.  I think the issue of why the public

 

      has the misconception that drugs are not safe can

 

      be tied directly to direct-to-consumer advertising.

 

                Many years ago, in my one opportunity to

 

      appear on the Today Show, I was interviewed by

 

      Debra Norville on the topic, and I was debating an

 

      industry representative, and I said that it would

 

      completely change the dynamics of prescribing and

 

      using of drugs in the United States, and I think it

 

      has.

 

                Two days ago, I was sitting at home

 

      watching TV, and for an hour and a half, every

 

      single ad on TV, every single ad was for a

 

      prescription drug, and it just has to have a

 

      dramatic effect on the way patients interact with

 

      their physician and how they get health care.  I

 

      think it was a mistake, but we can comment on that,

 

      too.

 

                Does anybody want to throw a few cents'

 

      worth in while we are prognosticating?

 

                                                                73

 

                MR. CLARK:  You mentioned something about

 

      E.R. Squibb challenging the world to meet his

 

      efficiency in his products that he manufactured.  I

 

      was just trying to point out that while he

 

      challenged the world, that challenge could prove

 

      fatal today, because today, Mr. Squibb or Dr.

 

      Squibb would be required to freeze his

 

      manufacturing technique, whatever it may have been,

 

      and that while his challengers came in with new

 

      techniques, he would be burdened with an approval

 

      process that would slow down his ability to

 

      compete, and we should be able to create a

 

      regulatory environment that protects the public as

 

      it still encourages innovation, and not just

 

      encourages the innovation for innovation's sake,

 

      but encourages applying it to the products and to

 

      improve the entire environment.

 

                DR. KIBBE:  Clearly, he couldn't do what

 

      he did then now, because the Federal Government is

 

      in his business now.

 

                MR. CLARK:  Exactly.

 

                DR. KIBBE:  And that has happened after

 

                                                                74

 

      World War II.  Before World War II, the Federal

 

      Government stayed out of everybody's business, and

 

      that is a dramatic change in the way we do business

 

      in the United States.

 

                We need to get to the desired state--I

 

      recommend Pennsylvania, far less hurricanes--the

 

      desired state, however, is going to be defined by

 

      Ajaz.

 

                   The "Desired State" of Science and

 

                     Risk-based Regulatory Policies

 

                DR. HUSSAIN:  I will do it from here.  In

 

      a sense, what we wanted to do, sort of build on the

 

      Manufacturing Committee discussion that was

 

      reported quite elaborately by Judy Boehlert, the

 

      chair of that committee, but to sort of now do a

 

      gap analysis, what we see as gaps between the

 

      current state and the desired state from an

 

      internal FDA perspective, what are the challenges

 

      we face internally, and get your feedback on that.

 

                So, what we have are three presentations,

 

      one, Helen will sort of look at the organizational

 

      issues, I will try to identify some of the

 

                                                                75

 

      scientific gaps, and Jon will identify some of the

 

      policy gaps and how we intend to sort of fill those

 

      gaps.

 

                If you could sort of give us feedback on

 

      are we missing in our gap analysis, it is a

 

      preliminary gap analysis right now, and then how we

 

      proceed, and then this will be followed by

 

      discussions and presentations by PhRMA and GPhA

 

      perspective on how they see the progress we have

 

      made and some of the challenges that remain.

 

                So, that is the discussion for this

 

      morning.

 

                DR. KIBBE:  That means you are passing the

 

      ball to Helen.

 

                DR. HUSSAIN:  Yes.

 

                DR. KIBBE:  Let's see how you follow my

 

      act.

 

                      Organizational Gap Analysis

 

                MS. WINKLE:  Believe me, in 100 years, not

 

      only could I not only follow your act, I wouldn't

 

      know where to begin, and my topic is so boring

 

      anyway.

 

                                                                76

 

                I am going to talk about organizational

 

      gap in the Agency right now as far as the desired

 

      state is concerned, and as Ajaz said, this is sort

 

      of a follow-up to some of the things we talked

 

      about at the Manufacturing Subcommittee, and I

 

      think it is really important that we look at these

 

      gaps and talk more about them, and sort of discuss

 

      how we can possibly fill some of them.

 

                I have some ideas on filling on some of

 

      them, but I think there is a lot more that we will

 

      need.

 

                DR. KIBBE:  There appears to be a gap in

 

      the computational problem, too.

 

                [Pause.]

 

                DR. KIBBE:  We are passing around a

 

      transportation note for people who need help to get

 

      to the airport.

 

                MS. WINKLE:  Is everybody leaving now?

 

      Gosh, you could at least have given me a chance.

 

                [Laughter.]

 

                MS. WINKLE:  As I said, I am going to talk

 

      about the organizational gaps and reaching the

 

                                                                77

 

      desired state, and I wanted to start off with, just

 

      a second, showing you the organizational chart of

 

      OPS, because I think as I talk about organizational

 

      gaps, you need to know a little bit about what the

 

      organization looks like, and I think you have a

 

      good idea, but I just wanted to point out we do

 

      have four offices.

 

                You actually heard from all four of those

 

      offices yesterday, but you say, in yellow, where

 

      the CMC is done in all four offices, so almost

 

      every part of the organization in some way is

 

      affected by the changes that are being made by the

 

      new paradigm and what we are trying to accomplish

 

      with the desired gap, which complicates the issues

 

      somewhat.

 

                It is very important as we look at the

 

      organizational gaps that it is multi-dimensional,

 

      it goes across all of the organization.  It is

 

      between organizations and it is within

 

      organizations.  There is lots of gaps here and we

 

      need to look at all of these gaps and figure out

 

      how we are going to handle them.

 

                                                                78

 

                It is outside of OPS and other parts of

 

      CDER.  We really do a lot of work with products

 

      with devices with CBER, so we need to be sure that

 

      those gaps are closed as we move forward in trying

 

      to accomplish the desired state.

 

                So, basically, what I am going to be

 

      talking about here is what we need to consider and

 

      resolve in our process or processes before we can

 

      adequately implement regulatory direction and

 

      support through applications process and review of

 

      what we are calling the desired state.

 

                I also want to point out as I talk, and

 

      you will see this a lot, that the organizational

 

      gaps that I am going to point out really intersect

 

      with the science gaps that Ajaz is going to talk

 

      about and the policy gaps that Jon is going to talk

 

      about, and you probably can't really address any of

 

      these separately although that is what we are

 

      making an attempt to do here.  But again as I go

 

      through the organizational gaps, you will see a lot

 

      of the intersections.

 

                What constitutes the gap in OPS and what

 

                                                                79

 

      are actually the process issues for implementing

 

      the desired state, and how we will review at

 

      different levels?  This is really some of what we

 

      need to talk about.

 

                One of the big problems here is the

 

      appropriate utilization and focus of available

 

      resources.  I am reading it wrong.  This is why I

 

      am having problems.  It is the resources.  We have

 

      a lot of human resources.  You have already heard

 

      some of the issues that we have had with how to use

 

      our best resources and how to focus those resources

 

      on those issues that are most important.  So, that

 

      is really one of the things that we have as part of

 

      the gap.

 

                We are not always focused on those issues

 

      which are the most important, and we don't always

 

      have the science expertise available to focus on

 

      the gaps or focus on the issues correctly.  So,

 

      this is a big gap that we have across the entire

 

      organization.

 

                There is a difference in products and

 

      regulatory requirements and review processes.  We

 

                                                                80

 

      are regulating ANDAs, NDAs, and BLAs, and BLAs even

 

      fall under a different act than the ANDAs and the

 

      NDAs.  So, there are some complications and some

 

      gaps there that we are going to have to look at and

 

      determine how best to handle.

 

                The organizational structure, the way it

 

      is set up really creates a really large gap in how

 

      we are going to move forward.

 

                I think we have made it clear in the past

 

      that in ONDC, I know Moheb has talked about this at

 

      different times, Dr. Nasr has talked about this at

 

      different times, that we have chemists from the

 

      Office of New Drug Chemistry that are located in

 

      the different clinical divisions, so that we lack

 

      consistency in how they make decisions often,

 

      because it is done outside of the whole chemistry

 

      structure, so to speak.  It is done within the

 

      Clinical Division, and we also lack the flexibility

 

      of being able to use our staff and to utilize the

 

      science and the staff because of these

 

      collocations.

 

                Actually, we have chemists in 18 different

 

                                                                81

 

      teams across the Clinical Divisions, and they very

 

      rarely interact with each other, so it really

 

      causes a lot of complications in how we do our

 

      work, and it will cause even more complications

 

      when we get into the new paradigm.

 

                I think one of the main gaps is that we

 

      are very process driven, not science driven.  This

 

      goes back to the earlier comment by Dr. Kibbe.  We

 

      are regulating a science industry.  It is a science

 

      industry that we are regulating through process.

 

                Some of the things that contribute to this

 

      is PDUFA in generic drugs, first in, first

 

      reviewed.  We have a tiered approach to our

 

      reviews.  We have heavy backlogs.  I think that

 

      Gary has made that point several times to this

 

      committee.  The workloads are big, the backlogs are

 

      big.  So, that is really driving us, too, to focus

 

      more on process than science.  So, this is causing

 

      us to really have to rethink how we want to do

 

      things.

 

                Part of what is adding to that workload

 

      and to the backlog is that we get too many

 

                                                                82

 

      supplements.  We require supplements on little

 

      changes that really have no significance in the

 

      manufacturing process.

 

                Also, part of the gap is the interaction

 

      with inspection.  We have a lack of appropriate

 

      reviewer involvement, and we get no feedback.  We

 

      do not get copies of 483s.  Once they have been in

 

      to the industry with the observations, we don't

 

      even have any correspondence in most cases with the

 

      inspection people on things that they find when

 

      they go out on inspections.

 

                So, how you are supposed to really have

 

      knowledge about the products that you are reviewing

 

      in the future or where you can use that knowledge

 

      that has been gathered and incorporate that into

 

      your thinking about reviews and products, you can't

 

      do, so that really creates a lot of gaps.

 

                One of the things that is going to create

 

      a gap in the future is the possibility of having a

 

      two-tiered system. As we talk about the desired

 

      state, as we talk about the things that are

 

      required under the desired state, we don't have

 

                                                                83

 

      regulations that are going to require manufacturers

 

      to submit pharmaceutical development information.

 

      We don't have regulations that are going to require

 

      them to do this thing or that thing, and in some

 

      places, I am not even sure we have the carrot to

 

      encourage them to do that.

 

                So, you are going to have some companies

 

      that are naturally going to submit this stuff, or

 

      naturally going to move toward PAT and toward other

 

      aspects of improving on their manufacture, but you

 

      are going to have companies that don't, so what we

 

      are looking at is the possibility of having a

 

      two-tiered system which is going to create a gap

 

      even within one reviewer.

 

                He is going to have to be able to look at

 

      both tiers and make decisions, and this is going to

 

      complicate issues a lot when we move ahead.

 

                We use guidances to accomplish

 

      consistency, and those guidances are sometimes very

 

      prescriptive, and this adds to the whole gap, and

 

      also not only are we using guidances for

 

      consistency, they are also up for interpretation. 

 

                                                                84

 

      Unless they are prescriptive, they are interpreted

 

      differently by different people, so obviously, we

 

      have some concerns about this.

 

                Organizational components are too

 

      reactive, and not proactive.  Now, this is caused

 

      by workload, and the workload continues to

 

      perpetuate the problem.

 

                You have to be reactive because you have

 

      so much work piled up in your In box that that is

 

      what you have to focus on, and it is very hard to

 

      be creative and innovative and think about those

 

      issues and problems that you are going to have down

 

      the road, think about, as Dr. Kibbe was talking,

 

      new therapeutics that are coming along or new novel

 

      delivery systems or different things like that, too

 

      busy moving the freight from day to day.

 

                Use of available scientific expertise and

 

      scientific collaboration.  Often within especially

 

      in ONDC, because they are broken up according to

 

      the clinical divisions, you may not have the

 

      necessary scientific expertise to look at an issue,

 

      to look at a problem, to know really what the right

 

                                                                85

 

      direction is for making a decision on a product.

 

                Also, we do not go out and use a lot of

 

      scientific collaboration.  I mean we have a lot of

 

      SGEs, we have several in this room that are helping

 

      us on different scientific issues of a broader

 

      nature, but we could be taking advantage of some of

 

      those and calling and getting more information in

 

      the future.

 

                There is a challenge in focusing on the

 

      appropriate questions or what are the right

 

      questions.  Reviewers have a tendency--and this is

 

      not any kind of negative against reviewers--but

 

      they do have a tendency to look at all the data

 

      that is provided, and we have not focused down on

 

      what the appropriate data is, and, therefore, the

 

      appropriate questions that we need to have

 

      answered.

 

                We have a lack of utilization of

 

      appropriate tools.  We could be using statistics

 

      more, all of us, to get better answers to some of

 

      the questions that we have around review.  There

 

      are other tools, as well, that we could be using

 

                                                                86

 

      that we are not.  One of the big areas I think that

 

      causes a gap is the lack of communication between

 

      disciplines, but I do want to add to this, there is

 

      also a lack of communication between organizations

 

      or components of the organization, and this is one

 

      of the things that we need to focus on to help

 

      close the gap.

 

                So, I did take a look at what we had done

 

      so far for closing the gap, because I think it is

 

      important to emphasize some of the stuff, because

 

      we do realize that we have some big gaps here.

 

                I do want to upfront say, though, that

 

      these are not all of things that we need to do.  I

 

      know that there is a lot more down the road that is

 

      going to come along, and I am really looking for

 

      advice from the Advisory Committee as to some of

 

      the things that we need to be thinking more

 

      carefully about, or make suggestions for some of

 

      the things that we could be doing to help close

 

      this organizational gap.

 

                One of the things we have been doing is

 

      making some structural changes in the organization

 

                                                                87

 

      In the Office of New Drug Chemistry, which Judy

 

      talked about yesterday for the Manufacturing

 

      Subcommittee, we are reorganizing the Office of New

 

      Drug Chemistry.  We are actually doing away with

 

      the collocation and making one Office of Chemistry

 

      when we move to White Oak.

 

                We feel that this is going to give us a

 

      lot of consistency or at least more consistency,

 

      and give us the opportunity to have more

 

      flexibility as to how we look at the review

 

      process.  We feel that this is going to have some

 

      real advantages to us.

 

                We are also, in our Office of Generic

 

      Drugs, we have set up a third division for doing

 

      chemistry.  The workload is so heavy in the office

 

      that we felt like if we had more divisions where we

 

      could spend more time and focus more on some of the

 

      issues, that we could help in some of the gap

 

      problems, having reviewers on inspections or as

 

      consultants to inspection, so have complete

 

      knowledge on products and the results of

 

      inspections.

 

                                                                88

 

                This is something that we have been

 

      working on.  We have been working with our Office

 

      of Compliance and with our field component.  We

 

      feel like we would like to have reviewers on

 

      inspection.  We think it is very important for them

 

      to go out and provide some of the scientific

 

      knowledge to the inspectors as the inspections are

 

      being done, but I don't think that part of the

 

      question even came up yesterday on resources to do

 

      this.

 

                This is a resource issue.  You are taking

 

      people away from their desk to go and--we have

 

      already talked about the workload being

 

      high--taking people away from the desk to go out on

 

      inspections and spend time away from their desks,

 

      but also this is costly, and like it or not, we are

 

      not flush with money, so we won't be able to do

 

      this in every inspection.

 

                But I do think it is important that before

 

      the inspections are done, even though the reviewers

 

      can't go out, that they provide consultation to the

 

      inspectors and talk about some of the issues that

 

                                                                89

 

      they have seen in the reviews of these particular

 

      companies and give them some advice on what they

 

      may want to focus on more in the inspections.

 

                One of the big things that is really

 

      necessary, in my mind, to closing the gaps, and

 

      again this sort of goes across the whole concept of

 

      the science gap and the guidance gap, and our

 

      policy gap, as well, is that we have a lot of

 

      questions we still need to answer and address.

 

                This is only a few of them, but I think

 

      there is a lot of things that we have not come to

 

      grips with on manufacturing science and how that is

 

      going to affect our review and what our review

 

      process is going to be to handle these things in

 

      the future - things like quality overall summaries.

 

                Dr. Nasr talks about incorporating this

 

      into the process of ONDC.  We have not come to any

 

      conclusions on this.  We are still in the proposal

 

      stage.  We need to decide, if this is the direction

 

      we want to go, what is the benefit of it to us, to

 

      the industry, and what we really need to see in

 

      that QOS.  So, that is a question that we need to

 

                                                                90

 

      look at.

 

                What we need in the way of pharmaceutical

 

      development information.  There is a lot of

 

      information that these companies have in the

 

      pharmaceutical development arena, and do we want

 

      all of that information.  If we get that

 

      information, what are we supposed to look at, what

 

      would we focus on.  We need to answer those

 

      questions, it is very important, before we start

 

      asking for this information.

 

                We have to have addressed that before, I

 

      think, companies are going to feel comfortable in

 

      providing it.  I think many companies see this as

 

      just more information they are sending us to look

 

      at and more questions they are going to get from

 

      us, so we really have to develop our processes.

 

                We also need to look at things like how

 

      industry will determine critical attributes, so as

 

      we look toward the desired state, that we are able

 

      to regulate that and include those in our process,

 

      and we need to know in all these cases what we will

 

      do as far as reviewing these.

 

                                                                91

 

                This is just a small part of the

 

      questions, I think, that we need to be addressing.

 

                Also, as far as closing the gap, we need

 

      to have a better understanding of what constitutes

 

      the design space across all products, and once we

 

      have a good feel about that, or understand that, we

 

      need to know when notification to FDA is necessary

 

      for change in manufacturing.

 

                We have not reached these conclusions yet,

 

      and we need to have working groups, whatever it

 

      takes, to really develop our own internal thinking

 

      on this and work with industry to make sure that

 

      the direction we are going is going to be useful to

 

      them.

 

                We need to have a better understanding of

 

      what risk for a product is and develop a systematic

 

      risk approach to review processes.  I keep seeing

 

      time and time again, people talk about risk

 

      management or risk processes, and stuff like that.

 

      I think when you talk about risk management, you

 

      are talking about something different for every

 

      person.

 

                                                                92

 

                I mean what is in my mind, what is in

 

      everybody else's mind in this room could be

 

      entirely different, and we at the Agency need to

 

      narrow down as far as review is concerned, decide

 

      what that means, how we are going to use it, and

 

      have a very, very concise program for ensuring that

 

      we do look at this in a fair and equitable way.

 

                Guidances.  Again, Jon is going to talk

 

      about guidances, but it is really necessary for us

 

      to go back. This will help us close the gap, but we

 

      need to go back and look at our guidance.  We have

 

      many guidances out there that are outdated, many

 

      guidances that are overly prescriptive, many

 

      guidances that don't fit into the new paradigm at

 

      all.

 

                Jon is in the process, he and his staff,

 

      of going through the guidances, removing some,

 

      redoing some, and looking at what guidances we will

 

      need for the future in order to incorporate some of

 

      the changes.

 

                Training.  This goes to the question Mel

 

      asked, training, training, training is necessary

 

                                                                93

 

      here.  We have so much to train.  We are doing some

 

      training, and I will talk about some of that, but I

 

      think it is really important and part of what we

 

      need to do to close the gap, is determine what

 

      really training our reviewers need and what

 

      training is necessary for the industry, and I don't

 

      think we have come to those conclusions yet.

 

                We need to determine how we are going to

 

      work under a two-tiered system if, in fact, that is

 

      the direction that we go, and we need to have

 

      developed the processes for doing that.  We need to

 

      develop an internal system for handling differences

 

      in Review Divisions.

 

                I met a couple of months ago with PhRMA on

 

      a RAC Committee on a dialogue session, and this was

 

      one of the things that came up was the need for a

 

      dispute resolution process, some kind of mechanism

 

      where, when there are differences in review, that

 

      we are able to handle those decisions and get

 

      information out as to how these issues are

 

      resolved.

 

                The last thing I have on here--and

 

                                                                94

 

      actually, I wrote this slide before we even had

 

      some of the conversations yesterday--was what is

 

      really important is we need to have appropriate

 

      metrics for measuring things.

 

                Today, in the review, we measure by what

 

      we accomplish, how many supplements we get.  Well,

 

      let me tell you when you are measuring supplements,

 

      and that is an indication of how good you are doing

 

      your job, you are going to want more supplements.

 

                We have got to really back off of the

 

      metrics that we currently have and look for those

 

      appropriate metrics to help close these gaps.

 

                So, some of the current steps we have

 

      across OPS, we mentioned before that we are setting

 

      up a working group under the Manufacturing

 

      Subcommittee of the Advisory Committee to begin to

 

      address many of these questions that we have.  We

 

      are also setting up some CRADAs to get some case

 

      studies to help us, too, in getting a better

 

      understanding of these issues and how we are going

 

      to handle them in our processes.

 

                ICH, too, is going to be an important part

 

                                                                95

 

      of helping us handle some of our organizational

 

      decisions especially Q8 as it looks at the desired

 

      state and implement some guidelines on it.

 

                We are doing some workshops.  We have a

 

      Workshop of Specification Setting and looking at

 

      how we need to have a mechanistic understanding of

 

      setting specifications in line with the direction

 

      that we are going.

 

                That particular workshop is set up in

 

      March, but I will be upfront with the fact that I

 

      think there is still going to be issues that come

 

      out of that workshop where we are going to have to

 

      look more specifically at some of the specification

 

      areas and really dig deeper into them, so I really

 

      anticipate more workshops than this just in the

 

      area of specifications, but I think a number of

 

      workshops are on the horizon in order for us to be

 

      able to address many of these questions.

 

                I actually think, too, one of the things

 

      that we are probably looking at having a workshop

 

      on is quality overall summaries.  If that is the

 

      direction we want to go, I think we need input from

 

                                                                96

 

      the industry and others, so that we have a better

 

      understanding of what we need for using that type

 

      of process and what it means to us in the industry

 

      when we do.

 

                We already have some collaboration with

 

      academics. As I said, we are involved in several

 

      CRADAs or in the development of several CRADAs.  I

 

      think these are going to be very helpful for us in

 

      getting a better understanding of some of the areas

 

      that we need to, or some of the questions we need

 

      to, answer, so that we can fill some of those gaps,

 

      and we have been doing some work with the Product

 

      Quality Research Institute.

 

                As I already said, we have an internal

 

      review of our current guidances, which is very

 

      important to helping us have the appropriate

 

      guidances out there.  We are developing a program

 

      for team interactions for inspections.

 

                We are sort of basing this on how we have

 

      handled PAT and the team approach, and we are

 

      working with Compliance in the field to develop a

 

      better way of handling inspections and including

 

                                                                97

 

      the Review folks in those inspections, and also a

 

      better way of getting the findings from those

 

      inspections.

 

                Training for reviewers.  We have already

 

      had a number of scientific seminars.  We have

 

      started that, especially, OGD has one every six

 

      months or so, and those seminars have been very

 

      beneficial to our staff in helping us have a better

 

      understanding of where we need to go, but we need

 

      more seminars and we need, again, really a set

 

      training program for all of our reviewers.

 

                We did form an OPS Coordinating Committee

 

      within the immediate office, and actually, Keith

 

      Webber and Gary Buehler are the chairs of that

 

      committee, and we will be looking at all the issues

 

      that come into OPS to try to ensure that we have

 

      consistency throughout all of OPS on handling

 

      these.

 

                One of the other things that we are in the

 

      process, I think, that will help with the gap is

 

      finalizing the guidance on comparability protocol.

 

                Also, in ONDC, as I said, we are really

 

                                                                98

 

      changing the organizational structure, but much

 

      more than changing the organizational structure, we

 

      are changing from a review program to an assessment

 

      program, and that assessment program will focus on

 

      quality attributes of the manufacturing including

 

      chemistry, pharmaceutical formulation, and the

 

      manufacturing process.

 

                So, the significant thing here is that we

 

      will be looking at much more the chemistry, the

 

      CMC, and that is where the assessment program is

 

      focused.

 

                We have the proposed QOS.  We have also

 

      implemented a team approach.  We are establishing a

 

      peer review process.  We have already done this on

 

      a limited basis to provide more scientific input to

 

      our scientists in their review processes, and

 

      helping everyone have a better understanding and

 

      sharing the knowledge that they learn from the

 

      reviews.

 

                We are implementing a Quality Systems

 

      approach. One of the things, too, that ONDC is

 

      doing, is they are developing a mock NDA under the

 

                                                                99

 

      new paradigm, under the desired state paradigm, so

 

      that they will have a better feel for some of the

 

      questions and issues that can come up, and they are

 

      looking at reducing supplement requirements.

 

                OGD has reorganized, as well.  As I

 

      mentioned, they have an additional Chemistry

 

      Division.  They are looking at changes in the

 

      supplement review and evaluation to determine if

 

      some of the supplements can be eliminated.

 

                They have also taken on the team approach

 

      on some applications, so that they have better

 

      utilization of scientific expertise and ensure

 

      consistency across similar product areas, and they

 

      are also looking at efficiencies in review to

 

      eliminate redundant or non-essential review

 

      activities.  So, they are very much involved, too,

 

      in some of the things that we need to be focused on

 

      in order to eliminate the gap.

 

                OBP, which is, of course, our newest

 

      review organization, is looking at supplement

 

      requirements to determine where we can eliminate or

 

      reduce supplements.

 

                                                               100

 

                Some additional steps.  I think we need to

 

      involve stakeholders in the review of guidances.

 

      Maybe under the Manufacturing Subcommittee we need

 

      a group that looks at some of the guidances.  I

 

      don't know how we need to do this, but I think it

 

      is a step we need to do.

 

                We need to determine how to handle the

 

      two-tiered approach, if we do it at all.  I have

 

      mentioned this before, and I think it is going to

 

      be important to involve industry and others in

 

      doing that.

 

                We need to have external workshops,

 

      develop a dispute resolution process.  One of the

 

      things, too, besides looking at regular GMP

 

      inspections, we really need to look at better ways

 

      to handle pre-approval inspection process.

 

                I would really like to see reviewers more

 

      involved in making some of the decisions on whether

 

      to do pre-approval inspections and to set better

 

      criteria for getting those done plus participate on

 

      the inspections if we feel they are necessary, and

 

      develop appropriate metrics.  These are things we

 

                                                               101

 

      haven't started on, but are obviously necessary,

 

      and I am sure there is others.

 

                Just to finish up, observations and

 

      conclusions.  I think we need to continue to

 

      address and analyze the organizational gap issues.

 

      I think they are going to be really important to

 

      us, to have determined what they are and to resolve

 

      how we are going to handle them in the future in

 

      order to move in the direction that we need to do

 

      to be able to regulate under the desired state.

 

                One of the things I think that is very

 

      important that we need to think about is culture.

 

      When I talk about culture, I am talking about the

 

      culture within FDA, and I am talking about the

 

      culture outside of FDA.

 

                There are a lot of changes that need to be

 

      here.  I realize that the culture is always a

 

      different area of thing to handle.  I thought

 

      Jerry's example was an excellent example of how the

 

      culture is a problem in some of the things that we

 

      are trying to do, and this is one of the things

 

      that we have got to manage and figure out how best

 

                                                               102

 

      to handle.

 

                All of this, all of this, the changes in

 

      the organizational gap will take time, and we need

 

      to be dedicating the time to make it happen.

 

                Also, as I said, a lot of this depends on

 

      resolving some of the science gaps that we have.

 

      We need to include stakeholders in making some of

 

      the decisions and developing some of the

 

      procedures.  We need to work closely, I think, with

 

      the stakeholders or we are really not going to have

 

      the answers that we need.

 

                The training of reviewers is important,

 

      and the thing I think that is going to be something

 

      that we have got to be open to is that as we move

 

      forward, we are going to see other gaps that we

 

      haven't anticipated, and we are going to have to be

 

      ready to fill those.

 

                That is all I have.  Thank you.

 

                DR. KIBBE:  Thank you, Helen.

 

                Should we take questions or you want to

 

      move to the--

 

                MS. WINKLE:  Let's go through all of it,

 

                                                               103

 

      yes.

 

                DR. KIBBE:  I will hold my really tough

 

      and incisive questions until later.

 

                        Scientific Gap Analysis

 

                DR. HUSSAIN:  Last night I had a phone

 

      call and I couldn't answer that, but this morning,

 

      at 7:00, I had another phone call from a company

 

      which recently got an approval for an inhalation

 

      product, and they were ecstatic. They had submitted

 

      a complete development pharmaceutics report, and

 

      that process went extremely well.  This was a

 

      one-cycle review approval for an important product

 

      including all the development report.

 

                So, I think that is a wonderful example

 

      that shows ONDC has already moved and things are

 

      moving in this direction already.  We probably will

 

      make a case study out of it, and so forth.

 

                Anyway, I would like to sort of focus on

 

      the scientific gaps.  I will use some background

 

      information.  I know a number of members on this

 

      committee who are new, so I thought I will spend

 

      some starting with the basics.

 

                                                               104

 

                I think, as Dr. Woodcock had come to the

 

      Manufacturing Committee, her articulation of what

 

      is quality has really come to almost fruition, and

 

      she is publishing this article soon.  The

 

      definition of quality is fundamental as we move

 

      forward, and there are some challenges as we move

 

      forward.

 

                Good pharmaceutical quality essentially

 

      means an acceptably low risk of failing to achieve

 

      the desired clinical attributes.  That is the goal

 

      of achieving quality.

 

                The challenge has always been, and you

 

      heard many of the discussions yesterday, saying the

 

      weakest link--and the weakest link is what we have

 

      to strengthen and address--how do we link

 

      measurements and risk?

 

                I think what we believe quality by design

 

      approach that we are developing under ICH Q8 is a

 

      way to help that. It is a multivariate model.  It

 

      is characterized during development.  You have to

 

      think about, when you think about quality by

 

      design, the clinical is a confirmation of that.

 

                                                               105

 

      That is the fundamental aspect that I think is

 

      going to be a significant challenge in how we

 

      achieve that.

 

                At the same time, you have to remember

 

      that development is only one part of it.  You

 

      essentially have to make sure you have a quality

 

      system.

 

                The final link between product and

 

      customer-driven quality attributes really means you

 

      have a good quality system for manufacture that

 

      brought us consistently also, that requires

 

      integrated product and process knowledge on an

 

      ongoing basis, because you don't stop learning at

 

      the time of approval.  In fact, you learn quite

 

      significantly after manufacturing status.

 

                You have to assure ongoing control, and

 

      you have to enable continuous improvement.

 

                In summary, I think Dr. Woodcock

 

      articulated this at our ONDC scientific rounds on

 

      October 6th.  The future definition of quality

 

      should be probabilistic in nature. That is the

 

      fundamental aspect, and we are not there yet.

 

                                                               106

 

                Science management, risk management, and

 

      quality management are critical aspects, and I

 

      think we really would like to be leaders in this,

 

      and I personally believe that we are.

 

                But let me take a look backwards from

 

      beginning with the end in mind.  Since we started

 

      the PAT Initiative, the cGMP Initiative, our focus

 

      has been on looking at the entire system, and we

 

      have been looking backwards from a manufacturing

 

      end to the entire discovery development product,

 

      and it is what do we learn from that.

 

                But before you look, before you measure,

 

      it is always good to make sure your measurement

 

      system is good, so you get your eyes checked.  That

 

      is the symbol there.

 

                The reason I was so sensitive to that is I

 

      think the dissolution variability from a

 

      manufacturing end, we really fully appreciate it

 

      when we are putting that white paper together, that

 

      today, companies don't have the ability to document

 

      lower variability than that of the calibrated

 

      tablet, and which is made with the conventional

 

                                                               107

 

      method, and so forth.  So, that was I think a stark

 

      reality that a lot of us fully understood during

 

      this process.  Art mentioned that, and so forth.

 

                So, what are the challenges here in the

 

      sense the challenge is organizational

 

      communication, and knowledge sharing and

 

      information sharing.  If you work in silos, the

 

      boundaries between organization, which I call

 

      interface, the quality of the interface between

 

      functional unit, that means the effectiveness and

 

      efficiency of the process, the interface can be

 

      handoffs between functions, and often is in need

 

      for better coordination, and that is what we

 

      learned through our GMP Initiative.

 

                The rapid and broad movement of

 

      information and knowledge sharing is necessary for

 

      process optimization between organizations, within

 

      any organization itself, so we have to move from

 

      technology transfer to knowledge transfer.

 

                But just toward the stage, reliability is

 

      a phrase that we often don't use in

 

      pharmaceuticals, but reliability has a very

 

                                                               108

 

      distinct body of information, body of knowledge

 

      associated with that.

 

                So, if you look at this figure, you have

 

      performance, you have life, shelf life, and you

 

      have a desired specification on both sides, on the

 

      performance.

 

                The first, Figure A is good performance,

 

      but poor reliability because the performance

 

      changes significantly over time, and the

 

      variability of the spec changes, too.

 

                The second one is good performance and

 

      good reliability over the life.

 

                The third is poor performance below spec.

 

                So, I think the key is when we think about

 

      performance, we are thinking about performance of

 

      the shelf life, the bioavailability, and so forth,

 

      remaining unchanged throughout the shelf life.

 

      Just to keep that in mind.

 

                In the current state, today, chemistry,

 

      manufacturing, controls, design information

 

      available in applications is limited and varied.

 

      Our reviewers have a high degree of uncertainty

 

                                                               109

 

      with respect to what is critical and what sort of

 

      process controls are necessary.

 

                Our reviewers have significant doubt on

 

      the level of process validation and process

 

      understanding.  So, they have no option but to

 

      focus on in-process and product testing.  So, in

 

      the pharmaceutical manufacturing from an

 

      engineering sense, testing is control, but in an

 

      engineering sense, control is very different.  It

 

      is a dynamic method. We don't do that.

 

                Risk coverage post-approval is a

 

      challenge, and supplements are a means for risk

 

      mitigation.  That is the way we have approached it.

 

                Traditional use of market standards--these

 

      are the pharmacopeial standards--as release tests

 

      are not effective for process understanding and

 

      continuous improvement.  In fact, by definition, if

 

      you have attribute data or so-called zero

 

      tolerance, continuous improvement is impossible by

 

      definition.  That is the definition in QS 9000,

 

      because we can only have continuous improvement

 

      when the product is already in spec.

 

                                                               110

 

                If you have zero tolerance criteria, by

 

      definition, the product is not in spec.  So, that

 

      is the fundamental thing.  Also, we understood and

 

      wrote about that in our Manufacturing Science white

 

      paper.

 

                We have variable test methods for physical

 

      characteristics, less than optimal systems

 

      perspective and approach, low efficiency and high

 

      cost of drug development and manufacturing, and

 

      continuous improvement is difficult, I would dare

 

      to say not possible.

 

                So, the success of the cGMP Initiative was

 

      to get a consensus desired state statement, so I am

 

      not using the exact words that we developed.  They

 

      are modified and the desired state statements

 

      adopted by ICH, these are the ones. Product quality

 

      and performance are achieved and assured by design

 

      of effective and efficient manufacturing processes.

 

                Since we are looking back from the

 

      manufacturing side, manufacturing goals are kept

 

      first.

 

                Product specifications based on

 

                                                               111

 

      mechanistic understanding of how formulation and

 

      process factors impact product performance, and an

 

      ability to effect continuous improvement and

 

      continuous "real time" assurance of quality.

 

                Now, let's start looking at this in the

 

      sense what are the gaps and how do you fill those

 

      gaps.

 

                Information and knowledge for regulatory

 

      assessment and decision process based on the

 

      desired state is information related to quality and

 

      performance and how the design impacts that.  So,

 

      we need to know impact of formulation and process

 

      factors on performance.

 

                We need information to judge and develop

 

      specifications based on mechanistic understanding.

 

      We need information to evaluate and facilitate

 

      continuous improvement, and continuous "real time"

 

      assurance of quality.

 

                The focus is on design, and if you are a

 

      formulator, especially one trained a few years ago,

 

      or if you have been in the design business, this is

 

      simply logical extension, so this is nothing new

 

                                                               112

 

      about this, but if you are not, then, you have to

 

      think differently the design process.

 

                Design is about doing things consciously,

 

      and not because they have always been done in a

 

      certain way.  It is about comparing alternatives to

 

      select the best possible solution.  It is about

 

      exploring and experimenting in a structured way.

 

      So, that is what design vocabulary brings to us.

 

                So, in the context of drug product

 

      development, design is about doing things

 

      consciously, so you start with the intended use.

 

      That is the fundamental issue.  You cannot forget

 

      the clinical use of the product that you are

 

      designing.  That includes route of administration,

 

      patient population, and all other things that

 

      impact on the intended use.

 

                That intended use defines for you what the

 

      product design should be.  You have options to

 

      select, and you select a product design.  That

 

      design leads you to design specifications, and

 

      those design specifications define the

 

      manufacturing process and its control necessary to

 

                                                               113

 

      develop those design specifications back to deliver

 

      the intended use.

 

                So, you have product performance, design

 

      specification that reliably and consistently

 

      deliver the therapeutic objective, and you have

 

      manufacturing capability, ability to reliably and

 

      consistently deliver the target for a design.  This

 

      is straightforward, logical, no rocket science, and

 

      we have been making and doing this for 100 years.

 

                So, that was the basis that we said we

 

      will develop the ICH Q8, and ICH Q8 document, which

 

      will go to Step 2 in Yokohama, I am confident about

 

      that, will essentially bring this type of

 

      information.

 

                It will not deal with the drug substance

 

      manufacturing part of it, but it will start with

 

      drug substance characterization.

 

                So, it will bring in characterization of

 

      components of drug product.  It will bring in

 

      aspects of manufacturing compatibility, and so

 

      forth.  Much of this information is sort of missing

 

      or varied in the current submission, and we are

 

                                                               114

 

      hoping, although the sections in CTD-Q (P2) are not

 

      ideal, we have to live with that because that is

 

      how everything goes in green, we felt that the

 

      sections provide enough room for bringing all the

 

      information to bear on that.

 

                So, we have made significant progress, and

 

      I think the draft 4 we are working on has captured

 

      most of this.

 

                What is the importance of design thinking?

 

      Design thinking makes the user paramount, ensuring

 

      that services we end up will do the job they are

 

      supposed to, as well as delighting the customer.

 

                Design thinking and methods provide new

 

      routes to better public services that meet people's

 

      needs and deliver value for money.  That is the

 

      key.

 

                We have been making tablets for 100 years

 

      or more.  It is a design problem.  We essentially

 

      have not used the vocabulary, we haven't brought

 

      that in.  Tools, such as pre-formulation

 

      characterization, and so forth, literally have

 

      become [inaudible], but that information often is

 

                                                               115

 

      missing in our assessment.

 

                So, if I distinguish between conventional

 

      and novel design for the sake of distinction in

 

      terms of how we use prior knowledge, the key aspect

 

      of this design and quality relationship is utility

 

      of prior knowledge.  For similar drug products, you

 

      have probably more prior knowledge, and for novel

 

      designs, you have to rely more on the experiments

 

      you generate, but prior knowledge is the key.

 

                If you are going to come with a new tablet

 

      formulation, and you have 300 similar tablet

 

      formulations on the market, how much more

 

      information do we need?  If you leverage the prior

 

      knowledge correctly and characterize your drug

 

      substance in a way to say all right, this is the

 

      way it is, you can leverage that knowledge.

 

                Level of mechanistic understanding will

 

      depend, will vary.  Pre-formulation programs, many

 

      good pre-formulation programs get to the mechanism

 

      of degradation, get to the mechanism of absorption

 

      including Bioform Classification System,

 

      characterization, that is the fundamental.  That

 

                                                               116

 

      defines literally every aspect of the manufacturing

 

      process and other things.

 

                So, if you have that information, if you

 

      will not be mechanistic completely, but you have

 

      valuable information, that moves forward.

 

                The challenge I think is to think about

 

      design during drug development.  As you develop

 

      your characterization and your development program,

 

      you have to keep in mind the ability to reliably

 

      predict performance, confirm as you progress.

 

      Every experiment you do next, say, a scale-up, is

 

      adding to your knowledge base, is a means to

 

      evaluate the predictability of your prior

 

      knowledge, and so forth.

 

                So, if you think about designing the

 

      entire development project from a design

 

      prospective, and capturing your predictability, you

 

      actually have an opportunity to move forward very

 

      quickly in terms of regulatory aspects, as well.

 

                So, level of understanding increases over

 

      time, and I think we have to recognize that.

 

      Structured empirical approach is often necessary

 

                                                               117

 

      because you often are not mechanistic.

 

                Use of prior knowledge to identify and

 

      select a design space for characterization is

 

      fundamental, and I think Ken Morris mentioned this

 

      yesterday.  People often jump into design of

 

      experiments without knowing what design space they

 

      want to explore.

 

                If you miss the prior knowledge, you

 

      actually increase your workload, you increase your

 

      cost by not being intelligent enough to say what

 

      are the critical variables upfront, and sort of

 

      exploring the design space.  You cannot approach

 

      this in a blinded fashion.

 

                For example, now, if you have multiple

 

      number of variables that you have to study,

 

      obviously, you cannot study all of them.  That is

 

      where risk comes in.  Prior knowledge and risk

 

      assessment is the way to address that, for example,

 

      failure mode effect analysis would be a means to

 

      say all right, these are the critical variables, at

 

      least these are potential critical variables, these

 

      are the ones we will select and move forward.

 

                                                               118

 

                So, initial conditions for screening

 

      experiments and then experimental conditions are

 

      then dependent on this prior knowledge and risk

 

      assessment.

 

                Impact of formulation and process factors

 

      on performance, why can't we leverage and be more

 

      intelligent about our clinical trial material

 

      itself, and how do we design clinical trials,

 

      because that is where the connection between

 

      quality and clinical comes together, and I will

 

      show you an example as we go.

 

                Similarly, with shelf life.  If you are

 

      getting mechanistic understanding, and so forth,

 

      prior knowledge and shelf life, I think is a

 

      wonderful opportunity which we don't utilize today.

 

                Just to give you an example, these are

 

      standard procedures in industry.  Here, is a work

 

      from Amgen in a sense how do they address the large

 

      number of variables as they go through process

 

      characterization, pre-characterization experiments,

 

      is to bring the prior knowledge to bear on this.

 

                So, process characterization studies start

 

                                                               119

 

      with pre-characterization work, screening

 

      experiments, interactions, and combinations of key

 

      parameters leading to process redundancy.

 

                They sort of covered that with a formal

 

      risk analysis.  So, these are standard procedures,

 

      and in many, many aspects, the formulation

 

      development process has built in robust approaches,

 

      but it is not formalized, it is not well

 

      understood.

 

                What is a robust design?  A robust design

 

      is not removing the source of variability, but

 

      designing a process or product to reduce the

 

      variability.

 

                A very simple example that in

 

      pharmaceuticals we have, is we know magnesium

 

      stearate is a wonderful lubricant, but it has a

 

      drawback of affecting dissolution, we know that.

 

                Half of the formulations that we have in

 

      our submissions actually have a robust design built

 

      in.  They will use a smaller model on sodium lauryl

 

      sulfate.  That negates the negative effect of

 

      magnesium stearate.  We have known that as

 

                                                               120

 

      pharmacists, formulators, and so forth, a long

 

      period of time, but we never captured that as a

 

      knowledge base.

 

                If you are making a tablet, you are

 

      compacting. Compaction has an effect on

 

      dissolution.  If you have right amount of

 

      disintegrating agent, you remove the effect of

 

      compaction force.  It's as simple as that.  That is

 

      what a robust design principle brings to bear on

 

      that.

 

                What is troubling often is, if you look at

 

      the SUPAC guidance, and if you look at the way we

 

      have regulated, the way we have done experiments

 

      often is to define our input or independent

 

      variables in terms of equipment.  Say, for example,

 

      if you look at the SUPAC guidance, we say equipment

 

      of same design and operating principles, you can do

 

      this, and so forth.

 

                That is not a quantifiable.  It is an

 

      identifier. So, we know that performance of a unit

 

      operation depends on material characteristics,

 

      particle attributes, equipment design, and

 

                                                               121

 

      operating conditions.

 

                Instead of sort of defining of input as

 

      equipment A, equipment B, and equipment C, and then

 

      doing a design experiment, if you are smarter, you

 

      will say all right, what are the forces acting on

 

      the particle irrespective of the equipment design.

 

      That removes that and improves your ability to

 

      generalize.  So these principles have been there

 

      for 60 years.

 

                Let me explain, in a sense, I think the

 

      key aspect here is risk-based specifications.

 

      Here, is an ICH Q6A decision tree.  Let me walk you

 

      through this.

 

                What specific test conditions and

 

      acceptance criteria are appropriate for a

 

      conventional or immediate release dosage form?

 

                Now, Professor Nozer corrected me before,

 

      so I will correct myself again.  He said this is

 

      not a decision tree, this is an event tree.

 

                Question 1 is:  Dissolution significantly

 

      affects bioavailability?  That is the Question 1.

 

                If the answer is yes, develop test

 

                                                               122

 

      conditions and acceptance criteria to distinguish

 

      between unacceptable bioavailability.

 

                But if the answer is no, you go down.  Do

 

      changes in formulation or manufacturing variables

 

      affect dissolution?

 

                If the answer is no, go down.  Adopt

 

      appropriate test conditions and acceptance criteria

 

      without regard to discriminating power, to pass

 

      clinically acceptable batches.

 

                The first question is how do you know

 

      dissolution significantly affects viability.  Most

 

      NDAs, not all, have a simple test that they do.  It

 

      is called a "Related bioavailability study."  They

 

      will compare a solution with a solid dosage form,

 

      and often you see they are superimposable.  That

 

      means dissolution is not rate limiting.  So,

 

      dissolution is not likely to affect that.

 

                Do changes in formulation variables affect

 

      dissolution?  Yes, all of them do, most of them do.

 

                If the answer is no, for heaven's sake, if

 

      you can find a formulation that doesn't have that,

 

      but you still put a dissolution test.  The question

 

                                                               123

 

      should be why, why do you need that dissolution

 

      test?

 

                So, some of the questions, how, why, and

 

      what really have not been addressed adequately, and

 

      our dissolution specification setting is one, two,

 

      three, these were your three batches, this is your

 

      specification.  Often, it is limited to that.

 

                I want to remind you that variability is

 

      inherent, and I did include a paper in your packet.

 

      This was published recently from Cambridge

 

      University and Pfizer.

 

                It said if you don't account for

 

      variability and you assume that meeting

 

      specification means you are bioequivalent, that may

 

      not always be true.  In fact, if you do this

 

      analysis, if your specifications are not set right,

 

      you have a 50-50 chance whether you are

 

      bioequivalent or not, or whether you meet

 

      specification or not.

 

                So specifications for dissolution are not

 

      likely to be the ones ensuring bioequivalence.  It

 

      is the entire control process that does that, but

 

                                                               124

 

      we focus so much attention on just one test, we

 

      miss the whole point.

 

                Let me come back to this decision tree.  I

 

      think in a quality by design thinking, this is what

 

      are my questions.  So, dissolution significantly

 

      affect bioavailability is a product design issue.

 

      You start with your pre-formulation, your biopharm

 

      classification, the solubility, permeability, and

 

      all that aspect, and you have an anticipation

 

      whether it will be affected or not, so when you do

 

      your related bioavailability study, you are

 

      conforming your past prior knowledge.

 

                So, postulate-confirmed based on mechanism

 

      or empirically, and that can apply to the question

 

      dissolution significantly affect bioavailability or

 

      do changes in formulation affect dissolution or

 

      not.

 

                But Jurgen points out, the key question is

 

      we have a mind-set 80 to 125, and that is the magic

 

      number.  Where did that magic number come from?  I

 

      think this is where the clinical relevance comes

 

      in, what is a relevant acceptance criteria to judge

 

                                                               125

 

      whether an acceptable bioavailability is there or

 

      not, and that is a clinical pharmacology question.

 

      That is where we link to the clinic.

 

                If you have a good PK/PD assessment, and

 

      so forth, you have far more information available

 

      to make a more rational judgment, and that is the

 

      question there.

 

                So, design of manufacturing and controls,

 

      and the question is how reliable those are, do

 

      changes in formulation and manufacturing variables

 

      affect dissolution? If the answer is yes, Are these

 

      changes controlled by another procedure and

 

      acceptance criteria?

 

                If the answer is yes, we come back and put

 

      a dissolution test.  My question is why?  If the

 

      dissolution test itself is variable, and so forth,

 

      why would you want to put another test?  You have a

 

      series of tests, and so forth, your chances of

 

      failure keeps increasing.

 

                So, the questions that we need to ask are:

 

      How good or how reliable are your design and

 

      controls that you have put in place for particle

 

                                                               126

 

      size, morphic form, and so forth, to address these

 

      conditions?

 

                So, overall risk-based CMC would ask why

 

      for these questions, but also, so what?  If

 

      dissolution is not rate-limiting, the question

 

      should be so what, why do we need a dissolution

 

      test, and so forth.

 

                So, this is how it all sort of comes out.

 

      So, quality by design thinking brings an overall

 

      CMC systems approach, for example, link to morphic

 

      form, particle size, stability failure mechanisms,

 

      and so forth, to address this in a systematic way.

 

                Continuous improvement is not possible

 

      today, because any movement is a change.  This is a

 

      direct cut-and-paste from our SUPAC guidance.

 

      Level 1 change, definition of change is this

 

      category includes process changes including changes

 

      such as mixing times and operating speeds within

 

      application/validation ranges.

 

                If you need to have validated those

 

      ranges, any movement within that is a change today.

 

      So, it requires to be reported.  If you change

 

                                                               127

 

      outside those ranges, you not only have to report

 

      it, but then you have to do a Case B dissolution,

 

      which is a profile comparison, and the supplement,

 

      and the stability, and so forth, so today, it is

 

      not possible.

 

                Our law and our regulations provide

 

      provisions for those approaches, and this is a

 

      Section 506A of the Act and 314.70 that we issued.

 

      We are required to make decisions based on

 

      potential to have an adverse effect on identity,

 

      strength, quality, purity, or potency of the drug

 

      product.

 

                We have used the phrases "substantial,"

 

      "moderate," and "minimal."  They are not very

 

      useful, they are not probabilistic, and I think

 

      that is where we have to work at.

 

                But also, if you look at CFR 314.70, there

 

      is a provision no change means no reporting beyond

 

      the variations already provided in the application,

 

      and that is where the design space comes in.

 

                So, what is this design space?  The design

 

      space is simply a space of knowledge or information

 

                                                               128

 

      where you know you will not affect your

 

      bioavailability, you will not affect your

 

      stability, and you will be in specification, but

 

      you are improving the manufacturing efficiency, you

 

      are improving the manufacturing process through new

 

      equipment, better controls through process

 

      adjustment in response to incoming input variables,

 

      and so forth.

 

                So, that is what continuous improvement

 

      is, and Box defined this years ago as evolutionary

 

      operations.

 

                So, that is how ICH Q8 information that

 

      brings reliability to your deliver design

 

      information, ICH Q9, which will develop the failure

 

      mode effect analysis and risk communication, too,

 

      all of them come together to define a design space

 

      for continuous improvement, and that design space

 

      will depend on the company's information that sort

 

      of comes about.

 

                You will know which area is the change,

 

      which area is not a change, and that is the map of

 

      Maryland, a weather map, so you shouldn't be in the

 

                                                               129

 

      red area.  That's about it.

 

                Yesterday, Steve showed this slide that I

 

      had developed for thinking about the entire system,

 

      how do you connect the dots.  I am not going to get

 

      into that, but I think the key aspect there is the

 

      knowledge space, and the knowledge space in

 

      relation to the clinical knowledge space and in

 

      relation to the manufacturing knowledge space all

 

      have to come together to sort of address this.

 

                A personal learning that I had going

 

      through the GMP process is a better appreciation

 

      for quality system.  I am still an academic at

 

      heart, and when you put me into a documentation

 

      mode, I get nervous, and great mounds of paper is

 

      something I want to avoid.

 

                The quality system that we have worked out

 

      in the GMP Initiative is actually quite nice and

 

      simple.  It says say what you do, do what you say,

 

      prove it, and improve it. Those are the fundamental

 

      principles.

 

                So, say what you do to FDA, is your

 

      pharmaceutical development information that you

 

                                                               130

 

      share with us?  If you say this is all I know, so

 

      that is what you are going to get.  If you say this

 

      is how much I know, and so forth, you get benefits

 

      from that, but then you have to do what you say

 

      consistently.  You have to prove it, and if you are

 

      unable to prove it, you have to ask why, and you

 

      have corrective actions.

 

                If you are unable to ask why, unable to

 

      answer why, then, there is a risk profile that

 

      increases.  And prove it is more optional, there is

 

      continuous improvement in innovation sort of comes

 

      in there.

 

                The challenges, I think in pharmacy, in

 

      pharmaceutical education, we have been doing this

 

      all along. What has been missing is a formal

 

      structure and communication tool.

 

                I draw some similarity here.  If I look at

 

      what has happened in chemical engineering, and now

 

      I think chemical engineering is going through

 

      soul-searching activities to redefine themselves,

 

      but this is how chemical engineering evolved.

 

                It started with industrial chemistry, unit

 

                                                               131

 

      operations, material and energy balance, chemical

 

      engineering thermodynamics and control, applied

 

      kinetics, process design, transport phenomena,

 

      process dynamics, process engineering.  Now, they

 

      are in molecular transformation, multi-scale

 

      analysis, and systems view.

 

                So, they went from industrial chemistry to

 

      unit operations, to chemical engineering science,

 

      to system engineering.

 

                Industrial pharmacy is still industrial

 

      pharmacy in the U.S., not as well in Japan, China,

 

      Europe, it's a pharmaceutical engineering degree

 

      literally.  So, we are still in that, and I think

 

      we can catch up on that, going to bring some of

 

      those principles.

 

                It is important to do that, it is

 

      important to bring a systems engineering

 

      perspective because not only we have to deal with

 

      the traditional goals of quality, the GMP

 

      Initiative offered new, non-traditional goals, that

 

      is, risk-based, flexibility, robustness,

 

      scalability, continuous improvement, innovation,

 

                                                               132

 

      and efficiency.  These are typically

 

      non-traditional goals.

 

                The characteristics of these goals are

 

      complexity and uncertainty associated with that.  The

 

      relationship between goals and characteristics

 

      that we are seeking is knowledge and information

 

      centric relationships.

 

                There are fundamental issues there,

 

      because if you don't get to this, our quality

 

      system will continue to be a paper chase exercise,

 

      and not really get to the heart of it, because we

 

      don't want to be lurching from fact to fact, from

 

      one quality system to another.  Unless this process

 

      is in the same sciences there, this will not

 

      happen.

 

                I will skip that and focus on where we

 

      are.  My assessment is this.  This is not rocket

 

      science, this is straightforward and simple for

 

      those who have been in this area for quite some

 

      time.  For those who are not, there is a need for

 

      education training.

 

                There are signs that I see.  The phone

 

                                                               133

 

      call this morning from a major company, and, in

 

      fact, I should have asked that I can share the name

 

      or not.  Their positive experience with the

 

      development report already in a four-cycle review

 

      is a good example that our folks can manage this

 

      process well, but consistency and making sure it

 

      happens consistently is a challenge.

 

                So, the immediate education need that I

 

      see going through the PAT training, and so forth.

 

      Now, for a broader training is introduction to

 

      statistical quality control.  That is fundamental.

 

      We are missing that, and I emphasize it is not

 

      biostatistics.  There is a distinction between

 

      statistical quality control and biostatistics,

 

      hypothesis testing, and where we keep missing that.

 

                I meet with the PhRMA Statistics Group,

 

      and so forth.  It comes back we are missing the

 

      quality dimension here.  We have to understand

 

      variability.  We have to focus and put training

 

      programs on molecular pharmaceutics and

 

      biopharmaceutics.

 

                We have gone to the molecular level in

 

                                                               134

 

      most of those areas.  Engineering principles is a

 

      key aspect.  Risk assessment and communication

 

      would be a program.  All of this will come together

 

      quite nicely with the ICH Q8/Q9 training program

 

      itself, but I think we would like to add some

 

      additional training.

 

                I know Ken has been working with us quite

 

      constantly on focusing on what the right questions

 

      are for the review process, but I think we can put

 

      a more formal training program on all of these

 

      aspects.

 

                I would like to say systems approach and

 

      thinking is important.  Unfortunately, most of the

 

      training programs that we go through, our BS/MS/BA

 

      programs actually takes us away from systems

 

      thinking to focus as narrowly as possible, and so

 

      forth, but in an applied area in the regulatory

 

      setting like this, systems thinking is important.

 

                Unfortunately, I go and talk about Deming,

 

      many people in the industry have never heard of the

 

      name Deming. I think we need to introduce people to

 

      Deming and others.

 

                                                               135

 

                Team building and communication will be

 

      the key.

 

                I will end my talk saying that coming

 

      together is a beginning, keeping together is

 

      progress, working together is success.

 

                The GMP Initiative brought us together.  I

 

      think the PAT Initiative took us further, and a

 

      smaller group is actually making progress.

 

                I am fairly positive.  I went through a

 

      quite depressive cycle in some of the challenges,

 

      and so forth, but I am fairly positive that I think

 

      we are on the right track, and we will achieve this

 

      rather quickly.

 

                          Policy Gap Analysis

 

                MR. CLARK:  I am going to deliver a talk

 

      about something of the policy gap, but what I will

 

      really be talking about is a guidance development

 

      process and some changes that we have done there.

 

      My talk will be quite pedestrian and quite short,

 

      which I hope to be some relief.

 

                I noticed in the agenda, at 10:30 we were

 

      supposed to break, but now it's after 11:00, and

 

                                                               136

 

      were this agenda an application, we would be found

 

      in violation of an agreement, and if we showed a

 

      pattern of being late, well, we might just be under

 

      a consent decree.  So, I think we are at some risk,

 

      so I will move us along and try to get us back on

 

      the path of righteousness, and such.

 

                I want to point out that somebody

 

      mentioned earlier today about failure, about

 

      failure data, I am sorry I didn't quite catalog who

 

      it was that brought it up, but the failure of data

 

      to point stayed in my mind.

 

                One of the things that we will be talking

 

      about in Yokohama in Q8 is what role that plays in

 

      an application, and to help define a design space,

 

      is there a place for us to use that in an

 

      evaluation of an application, and if you have

 

      determined where your system fails, can that offer

 

      you some relief as to where you operate.  I wanted

 

      to just bring that point up as I start in this

 

      speech.

 

                In the GMPs for the 21st Century, some CMC

 

      guidance documents are out of synchronization with

 

                                                               137

 

      that rollout that you all have seen by now, but the

 

      guidance process that developed these documents has

 

      strong and weak points, and one of the main

 

      strengths of this is the technical input from our

 

      staff, from our review staff.

 

                One of our weaknesses is in the

 

      decisionmaking process for actually moving the

 

      documents from step to step and getting them out,

 

      which causes it to be very slow.

 

                I would like to really dwell on the

 

      strength.  One of the things that we need to take

 

      away from our previous guidance development process

 

      is that these deliberative processes are well

 

      meaning, and these people are highly trained, and

 

      they are experts at what they do.  At every step,

 

      they are trying to articulate the things that are

 

      on their minds and how to get applications approved

 

      in the best way.

 

                They may have become proscriptive and

 

      prescriptive, but that is not a failure in their

 

      attempts to articulate the best way to get an

 

      application approved.

 

                                                               138

 

                We believe that there may be a better way

 

      to articulate that point, and obviously, with the

 

      rollout, and you compare the rollout to the

 

      documents that we have on our guidance page, there

 

      is the gap, and most people know that, that are

 

      familiar with the two sets of documents, so I will

 

      move on from there.

 

                The draft cycling that was the weak point,

 

      I will point out this is the old draft cycling, and

 

      you will see that there was a CMCCC working group

 

      assigned from a CMCCC committee body.  That CMCCC

 

      would define a group to work, and we will call that

 

      the body for now, to develop a document.

 

                They would go ahead and develop a

 

      document, and this might take six months, and it

 

      might take two years, and it might take five years,

 

      but they would develop an articulation of the areas

 

      of interest for that document.

 

                They would then proceed to take that and

 

      go through each review team, through some kind of a

 

      hierarchical structure in the organization.  Those

 

      review teams would then have comments, not unlike

 

                                                               139

 

      the public comment system, and those comments would

 

      go back to that review group, and they would

 

      redraft the document, which might take another year

 

      or two.

 

                Because these people are not dedicated to

 

      that task, they are also reviewing drugs, they are

 

      also involved in a lot of other efforts like ACPS,

 

      and they are also involved in guidance development.

 

                So, they go back to the review teams, goes

 

      back to that CMCCC body, and then it goes back up

 

      to the working group or back up to the committee

 

      for review, and then from  the committee, it goes

 

      to an OPS editor.

 

                Now, that process, those steps might take

 

      as much as six months, it might be a year.  The OPS

 

      editors then have a go at making sure that the

 

      legal language is current with the desires of our

 

      legal staff, and they might pass it on to the legal

 

      edit if their suggestions are minimal, and so on.

 

                If not, if the suggestions are strong and

 

      get into the body of the document to a large

 

      degree, it might actually go right back up to the

 

                                                               140

 

      body and have to go all through all that stuff I

 

      just mentioned all over again, and were I

 

      mean-spirited, I could go through it a second time,

 

      but I won't.

 

                Well, you go to the legal edit, then, it

 

      goes out to public comment.  Then, you have public

 

      comments dockets come back.  You might have 1,000

 

      comments if you are lucky. It might be a couple

 

      inches thick if you are lucky, and it might be a

 

      foot high if you are not so lucky.

 

                If that happens, well, it will happen,

 

      then, you have to catalog all those comments,

 

      address each of them somehow, address them by

 

      groups or individually, and then if you have to

 

      make substantial changes to the document in order

 

      to address those comments, go back up to the top,

 

      and if I were extra mean-spirited, we could go

 

      through this whole thing again.

 

                I think you can understand that that could

 

      be a laborious process, and that is my excuse for

 

      why guidances take so long to get out of the

 

      Agency.

 

                                                               141

 

                When somebody says that a guidance will be

 

      available soon, they may think it is going to be

 

      available soon, because they think they are near

 

      the end of the process, or what they don't maybe

 

      not understand is that there is an iteration that

 

      they hadn't predicted.  So, that is something we

 

      have been dealing with over the years.

 

                This is a slide that puts into words some

 

      of what we just discussed, but it also points out

 

      that there is a rapid change in FDA thinking over

 

      the last couple of years. It leaves slower efforts

 

      to catch up.  The guidance development is, in fact,

 

      a slower process.

 

                There is an investment of time, and the

 

      documents may be slow, but they have a lot of

 

      momentum.  You have that many people working that

 

      hard, that many smart people working that hard over

 

      that many years, and the document gets momentum.

 

                The guidance content and the new direction

 

      are managed actually by two different groups.  We

 

      have this OPS policy direction setting group, which

 

      had some involvement from the reviewers and staff,

 

                                                               142

 

      but the guidance content was being managed by the

 

      review staff directly with reciprocal input from

 

      the higher end, the OPS level.

 

                We have taken some actions to try to

 

      remedy the situation.  We have moved coordinating

 

      and decisionmaking from the CMCCC working group to

 

      an OPS office level group, and have created the OPS

 

      CC.  Helen mentioned that a little earlier.  They

 

      have a lot of tasks, one of which will be to help

 

      us coordinate these guidance documents.

 

                It will move guidance content management

 

      up to OPS with some lookback down to the review

 

      level when we need that expertise that is at the

 

      review level.

 

                We disbanded the CMCCC as such, as words.

 

      Of course, we have the same people wearing hats

 

      with new letters on them, OPS CC, but it has a

 

      different function, and I will show you that in the

 

      next slide.

 

                The mandate through OPS CC is to recruit

 

      technical input from scientists when we need it.

 

                The new process looks something like what

 

                                                               143

 

      I have on the board now, where you have OPS CC

 

      actually managing the creation of the documents,

 

      getting science input from selected teams, getting

 

      input back to OPS CC.  Then, you have a smoother

 

      process on the high end of that document formation

 

      process.  So, we hope that that will shorten things

 

      up a bit up there.  It might take 10 or so years

 

      off the process, which should improve things.

 

                The OPS edit and the legal edit have never

 

      really been hugely time-consuming except that they

 

      can cause reiterations, but it is that iteration

 

      that we have tried to smooth out.  Of course, the

 

      public comment isn't changing. We still have the

 

      dockets and dockets management.

 

                We are trying to synchronize the effort,

 

      but there are a number of techniques we intend to

 

      use to synchronize the effort of guidances with the

 

      rollout of the two-year effort in the GMPs for the

 

      21st Century.

 

                One is, of course, obviously, a revision

 

      of drafts.  We have drafts out there.  We have

 

      public comments that are available for many of

 

                                                               144

 

      those drafts, bring those in, and we might actually

 

      put out for a second draft some of these documents,

 

      because we may incorporate public comment, we may

 

      incorporate some of the new thinking, and if the

 

      document is substantially changed, we may not go

 

      from draft to final.  We may go from draft to

 

      public draft to get more comment.

 

                Another option is the withdrawal of

 

      documents.  I would see that option mainly for

 

      finalized guidance documents that just have become

 

      obsolete, and perhaps no longer do any good for us,

 

      so we may actually consider withdrawing some of

 

      them.  We have done this in the past, and we may

 

      seek to do it in the future.

 

                Another effort is enforcement discretion.

 

      We may actually use guidance to bring into line

 

      more of our practice where sometimes a practice has

 

      found its way into regulation, where the regulation

 

      will require you to do something that we really

 

      don't necessarily believe is constructive at this

 

      point anymore, and we may use guidance to iterate

 

      an enforcement discretion over some terms to

 

                                                               145

 

      address some of the regulation requirements.  That

 

      would pretty much be viewed as a precursor to

 

      actually revising the regulation itself.

 

                There is also a consideration of options

 

      in OTG other than guidance.  The question and

 

      answer format where we have a simple question and

 

      we want to answer it with a paragraph and

 

      hopefully, it doesn't turn into a chapter, we post

 

      it on public Internet, try to keep it simple.

 

                It would go through our guidance

 

      procedure, but hopefully, be much quicker, and not

 

      try to address a large range of things, but just to

 

      keep it to one topic, keep it simple.

 

                We also want to look into a Manual of

 

      Policy Procedures, to expand that.  I don't believe

 

      that we have actually exploited our Manuals of

 

      Policy and Procedures enough to articulate what we

 

      would like our internal staff to be doing, have it

 

      in an if/then format, directed toward OPS staff.

 

                What is nice about the Manuals of Policies

 

      and Procedures is that they are publicly available

 

      once we are done, and they are rapid.  They can be

 

                                                               146

 

      rapid to get out, and we have more flexibility in

 

      what we say in them.

 

                Back to guidances in particular.  We

 

      really would like to have a more risk-based

 

      approach expressed in the guidance, reverse recent

 

      tendency toward proscription and prescription,

 

      which I think that if anyone has read some of the

 

      recent drafts, you can see there is a difference

 

      between what was published in 1987 and what we have

 

      been publishing in recent times, and that they tend

 

      to be much more detailed and much more instructive

 

      as to exactly what you should be doing.

 

                We try to get up to a higher level and try

 

      to rely more on the science for the specific issues

 

      to drive the decisions that are made.

 

                The manufacturer should choose the

 

      technology and the approach to problem solving

 

      where we should be evaluating more of the science

 

      behind whether or not the quality, the ongoing

 

      quality is short, and as Ajaz put up there earlier,

 

      what is the acceptably low probability of not

 

      meeting the clinical intent.

 

                                                               147

 

                Focus efforts on assurance of reliable

 

      product quality, and not on technology.

 

                Input into our processes is one of the

 

      things I would like to propose here, is for this

 

      body to seriously consider, creating a fact-finding

 

      group to help us with guidance processes.

 

                Is there a potential gain from formation

 

      of this fact-finding group to help us determine how

 

      we move forward to the desired state?

 

                Some of the questions that fact-finding

 

      group would be asking or asking and answering would

 

      be:  Do we need all the guidances that we have now?

 

      Where are they incongruent?  Provide advice as to

 

      what guidance industry needs as we work toward a

 

      new regulatory paradigm, and what should be the

 

      prioritization of those documents if we have any of

 

      them at all.

 

                So, I would like to propose that this body

 

      be engaged in that effort at some level to be

 

      deciding who would be on that body and, of course,

 

      it would be within the rules of the advisory

 

      committee system.

 

                                                               148

 

                That is the end of my talk.  Since I am

 

      the last of the three, I guess I will stay at the

 

      podium and be the target for the questions.

 

                DR. KIBBE:  It depends on how much time

 

      the other two speakers will need and how much time

 

      we allow for questions.  We can always come back to

 

      the topic after the lunch break, but if there are

 

      some quick questions, we can handle, Nozer?

 

                DR. SINGPURWALLA:  Not quick.  I have lots

 

      of questions for Ajaz, and I am sure he expects

 

      those.  So, perhaps we should postpone them until

 

      another avenue, or I can privately communicate.

 

                DR. KIBBE:  No, we will get to them.  I

 

      want them on the record.

 

                Go ahead.

 

                DR. MEYER:  Just one quick comment.  I

 

      like the idea of a question and answer format in a

 

      public Internet, frequently asked question kind of

 

      thing.  I think that would save the Agency some

 

      time.  It will be easy for the sponsors.  You won't

 

      have to deal with repeat questions, so I think that

 

      would be a step forward to have that.

 

                                                               149

 

                MR. CLARK:  Thank you, Committee, thank

 

      you, Dr. Kibbe.

 

                DR. KIBBE:  Meryl, go ahead.

 

                DR. KAROL:  I have heard throughout this

 

      morning about the risk-based approach towards your

 

      changes, and I wondered what do you have in mind

 

      for training and education of the personnel in

 

      order to make them knowledgeable about risk-based

 

      approaches?

 

                MR. CLARK:  The training of the personnel

 

      is really an ongoing thing.  It isn't really a here

 

      it starts and here it ends.  We have the people

 

      involved.  We have a peer review system that Moheb

 

      has set up where they come together after they have

 

      finished a review, and they present it to their

 

      colleagues.

 

                We, from the Policy Groups, we go in and

 

      we listen.  We have comments.  We steer them, we

 

      try to steer them toward the things that have been

 

      done well, and to bring that out.  A lot of the

 

      practices that are involved in a review of an

 

      application are, in fact, risk-based practices, and

 

                                                               150

 

      sometimes a reviewer just doesn't bring that up to

 

      the surface and make that well known.

 

                Part of our role in that particular venue

 

      is to point that out, to say, well, you took a

 

      risk-based decision here, we would just like you to

 

      bring that up to the surface and make it the theme

 

      of the document as opposed to going down into the

 

      technology and making that the backbone.

 

                So, it isn't really a big stretch for

 

      these people to grasp onto it.  They are all very

 

      highly trained.

 

                We do have training programs in mind.  We

 

      haven't really implemented a lot of them upfront

 

      yet, but the goal is to have a top-down approach.

 

      Dr. Morris has been involved at some level at

 

      helping us train managers into the thinking and to

 

      work that down through the management levels.

 

                We have gotten down to the team leader

 

      level at their last presentation, but the rollout

 

      did take up all our resources up until just now.

 

      So, we will be re-initiating that effort.

 

                DR. KIBBE:  Jurgen.

 

                                                               151

 

                DR. VENITZ:  A few comments, one more

 

      fundamental comment to Jon's presentation that has

 

      to do with my favorite word, and that is risk.  You

 

      talk about how risk is being assessed, and you have

 

      emphasized primarily the probabilistic component,

 

      that you have tried to predict the likelihood of

 

      something happens.

 

                Well, risk has a second component, and

 

      that is a judgment, how bad is what you measure, or

 

      how potentially bad can it be.  I think one of the

 

      things that you will face is within the Agency, is

 

      this culture of always assuming the worst, anything

 

      that happens we can assess whether that happens

 

      often or not is bad.

 

                Well, risk really requires you to make a

 

      judgment as to how bad or how not as bad it might

 

      be.  So, I am just pointing that out, risk is not

 

      just measuring probabilities, but it is also

 

      assigning utility values to those probabilities.

 

                Two small comments to Helen's

 

      presentation.  I would encourage, especially with

 

      all the changes that are occurring, that your staff

 

                                                               152

 

      participate in all those industry meetings,

 

      particularly the pre-IND and the end of Phase II

 

      meetings, because I am pretty sure that is where a

 

      lot of things are being discussed that are relevant

 

      to the change in GMP and PAT, and what have you.

 

                As far as the briefings are concerned, you

 

      might consider something like a question-based

 

      review where not the entire material gets reviewed,

 

      but you are actually focusing in on things like

 

      source variability on things that are potentially

 

      at risk.

 

                DR. KIBBE:  Thank you, Jurgen.  What we

 

      really were hoping for is if you had a quick

 

      question, because I want to get the next two things

 

      out.  If you have got a long discussion, we will do

 

      it right after lunch.

 

                Let's go ahead and get Mr. Ahmed to talk

 

      about the FDA Critical Path Initiative, a generic

 

      industry perspective.

 

             Generic Pharmaceutical Association Perspective

 

                MR. AHMED:  Good morning, everybody.

 

                Sorry for this delayed presentation.  I

 

                                                               153

 

      know a lot of you must be looking for a break, but

 

      just bear with me.  It's not that bad.  It will be

 

      fairly short because I am presenting from a generic

 

      perspective, and clearly, the Critical Path

 

      Initiative does not really look into the generic

 

      industry the way other presentations or the

 

      Internet on the FDA web site.

 

                First of all, I would like to thank Helen

 

      and everybody at the Agency for providing GPhA the

 

      opportunity to present their view regarding the

 

      Critical Path Initiative.

 

                Gordon actually asked me to do this

 

      presentation on behalf of GPhA.  I am from American

 

      Pharmaceutical Partners, which is a direct

 

      injectable manufacturer.  We have a proprietary

 

      drug under review right now at the FDA.

 

                What I am going to talk about, first, is

 

      what is the overview of generic industry, what part

 

      the generic industry really plays in the health

 

      care system in the United States, the Critical Path

 

      Initiative concept, the benefits of the Critical

 

      Path Initiative to both the generic industry, as

 

                                                               154

 

      well as the innovator companies.

 

                I think one of the most important

 

      component of the U.S. health care system is generic

 

      industry, as you can see. Fifty-one percent of the

 

      prescriptions dispensed in the United States were

 

      generic drugs.  This data is based on the 2003 IMS

 

      data.  I am sure the utilization of drugs will

 

      increase as time goes by.

 

                About 8 percent of overall cost of

 

      prescription drugs is contributed by the generics.

 

      There are several off-patent products which still

 

      don't have any generic competition, and that is

 

      really hurting the consumer in a lot of respects,

 

      because these products have been off patent for a

 

      while, and I will elaborate on those as I go on.

 

                When we reviewed the Critical Path

 

      Initiative, we found that it really provides

 

      significantly improved tools for evaluating

 

      basically safety, efficacy, and characterization of

 

      the drug substance like the product/manufacturing,

 

      because I think this is a very bold concept

 

      proposed by the Agency, because over the last

 

                                                               155

 

      15-plus years I have been dealing with the Agency,

 

      this is really a breath of fresh air, because I

 

      still hear that titration is a better method than

 

      is HPLC, because it's in the USP, so this is really

 

      very good thinking on the part of the Agency.

 

                We have got to get out of that mode,

 

      because, on the one hand, we hear from FDA good

 

      science, good science, good science.  We file an

 

      application and provide a better method to the

 

      Agency.  There is now we like potential metric

 

      titration because in the USP, your HPLC method or

 

      LCMS method is not good.

 

                So, I think this is a good shift.  At

 

      least from our standpoint, we think that science

 

      really holds a lot more promise and can hoe a lot

 

      more ground than regulations.

 

                We like FDA's approach.  We like the

 

      collaborative approach.  I think it is important

 

      that we involve the academics, the patient groups,

 

      the industry, as well as the regulatory bodies,

 

      because really at the end of the day, all of us

 

      have the same function, which is to protect the

 

                                                               156

 

      American public and to provide a service, because

 

      the drugs used by folks is really for people who

 

      are unfortunate, because they have to use those

 

      drugs, so we have to make sure that we provide the

 

      highest quality product, which is the most

 

      efficacious and has the lowest side effect.

 

                The desired outcome meaning yes, faster

 

      approvals. Again, a little bit of a misnomer

 

      because just to give an example, we filed for a

 

      CB30 to extend expiration dating for a very

 

      critical product.

 

                It took FDA 60 days to first acknowledge

 

      that we filed the CB30, and then it will be another

 

      six months before we make a decision, so what is

 

      the point in filing a CB30?  That is what I really

 

      wanted to bring to Helen.  If we want the desired

 

      outcome to be faster, let's please be more

 

      pragmatic, because if it is built around the system

 

      the way it is right now, the desired objective is

 

      really very far away.

 

                Again, the primary emphasis is on

 

      discovery, and it really helps pharma, and clearly,

 

                                                               157

 

      we are a big proponent of that because what pharma

 

      produces eventually has the opportunity to become a

 

      generic, so we are imploring the Agency to work

 

      with pharma to enhance drug development process and

 

      give the pharma folks faster approvals.

 

                From a generic industry perspective, I

 

      think the Critical Path Initiative is also very

 

      helpful, and I request Helen and Ajaz and everybody

 

      else to really think it hard because the generic

 

      industry represents a big opportunity for the

 

      American health care system, as well as for the

 

      Agency itself.

 

                We would like to be part of this

 

      initiative because there are lot of opportunities

 

      where we can minimize the pains we go through on a

 

      daily basis, as well as you folks.

 

                Again, the goal here is to have timely

 

      submissions as well as timely approvals, so the

 

      consumer can benefit.  As you know, the prices,

 

      when the product becomes generic, almost within the

 

      three months, the price of the generic drug is

 

      almost 90 percent less than the innovator product,

 

                                                               158

 

      so very clearly it helps the American public

 

      significantly from a financial standpoint.

 

                Again, we very earnestly request the

 

      Agency to really look into providing guidance to

 

      MDIs.  As all of you know, it took seven years for

 

      Albuterol to become generic. We don't want to go

 

      through that scenario again.  There are still a lot

 

      of products which there is no guidance for.

 

                The products, especially the transdermal

 

      products, which have no guidance, the inhalation

 

      products, there is still no guidance, and also,

 

      again, I don't want to take anything away from

 

      Ajaz's PAT Initiative, but in the entire PAT

 

      Initiative, there is no mention of sterile

 

      products, and currently, in the United States,

 

      about 20 percent of prescription drugs are sterile

 

      products, which includes parenterals, which

 

      includes inhalation products and topical products.

 

                So, I would really again urge FDA to look

 

      into how they can help the sterile product

 

      manufacturers in terms of providing guidance and

 

      reducing regulatory burden.  Clearly, we are

 

                                                               159

 

      looking for better collaboration with FDA.  We

 

      would like to be partners with FDA in every respect

 

      we can, but again it's a two-way street.

 

                Sometimes we get a lot of resistance from

 

      OGD in terms of pestering them too much or in terms

 

      of arguing on scientific issues, so we really would

 

      like to have a more collaborative effort.

 

                As part of GPhA, I would really urge FDA

 

      to look into it in terms of what our objective is,

 

      to make the safest product, but apparently there

 

      are some scientific disagreements at times, and we

 

      want to minimize the pains we go through in terms

 

      of the review process.

 

                Lastly, I would like to talk about the

 

      21st Century Pharmaceutical GMP Initiative.  I

 

      think it's a step in the right direction, but there

 

      are a lot of issues we really need to look very

 

      hard at.  One of the issues is inspection.

 

                There is so much inconsistency in the

 

      inspection process, and to give you an example, an

 

      inspector going to a sterile manufacturing facility

 

      in New Mexico really does not even cover one-tenth

 

                                                               160

 

      of what is covered in the Chicago District.

 

                So, like we are from the Chicago District

 

      and we get the highest scrutiny, I can tell you

 

      that for a fact.  Susan Bradley basically really

 

      keeps us on our toes, but there is so much

 

      inconsistency as I go from district to district.

 

      Please look into this because this really hurts the

 

      generic manufacturers especially because of the

 

      patent issue, and things like that.

 

                The other think is I keep on hearing the

 

      Science-based Initiative, Science-based Initiative,

 

      but we are still getting questions from the

 

      reviewing chemists, which is really a total waste

 

      of time for us and you both, the reason being that

 

      we cannot change basic science, and nobody can

 

      change basic science.

 

                But to give you an example, not too long

 

      ago I got a request from a reviewing chemist asking

 

      me to provide a chromatogram for mannitol where the

 

      quantitation was 229 nanometers, and I picked up

 

      the phone and called the reviewing chemist and I

 

      said, "Please, mannitol has no pi bonds, they are

 

                                                               161

 

      all sigma bonds," and I [inaudible] off any sigma

 

      bond which the electrons could be transferred to

 

      sigma star at 220 nanometers."

 

                He said, "I still want to look at the

 

      chromatogram."  So, please, please be open-minded,

 

      please listen to the folks who really do this for a

 

      living.  They know the science.  They would not

 

      present data which does not make sense.

 

                So, that is part of the 21st Century

 

      Pharmaceutical GMP Initiative.  Let's be

 

      open-minded, let's discuss with the industry,

 

      because we folks, we put a lot of time and effort

 

      in terms of manufacturing batches, testing batches,

 

      and we want to produce the highest quality product

 

      and to have the minimum risk.

 

                However, we need to be very open-minded.

 

      We need to depend on science rather than

 

      regulation.

 

                Thank you.

 

                DR. KIBBE:  Judy has a question.

 

                DR. BOEHLERT:  I have a comment.  I am

 

      going to help you out.  You said you had a problem

 

                                                               162

 

      with USP methods, titrations.  Well, I have several

 

      solutions for you.

 

                First of all, USP would love to get your

 

      HPLC method.  Second of all, USP allows the use of

 

      alternate methods, so as long as your HPLC method

 

      is equivalent to a better titration, you may use

 

      it.

 

                Now you have got the FDA to deal with.

 

      They are talking about risk, and they are talking

 

      about science.  I would hope that the FDA would

 

      look at a submission favorably if you have

 

      demonstrated that your HPLC method is equivalent to

 

      the titration method and the compendium, and you

 

      can go forward.

 

                I know you probably do get questions along

 

      those lines, but I think we need to move past that

 

      and ask the right questions.

 

                MR. AHMED:  Judy, what happens is that

 

      they say yes, HPLC method is better assurance than

 

      equivalence data, so what is the point?  If you are

 

      showing equivalence data, then, what is the point

 

      of using HPLC method?

 

                                                               163

 

                DR. BOEHLERT:  Well, because you don't

 

      have to do the titration, and you really do get

 

      better data with the HPLC method.

 

                MR. AHMED:  That is what my point is.

 

                DR. BOEHLERT:  Titration might be a great

 

      method for releasing product, that you might want

 

      to use HPLC particularly during development to

 

      learn more about your product.

 

                MR. AHMED:  And like we are providing a

 

      stability indicating method, and we are telling the

 

      reviewing chemist, please, titration cannot predict

 

      the stability indicating nature of the product, and

 

      still we are asked to provide comparative data.

 

      So, that is what my point is.

 

                DR. BOEHLERT:  I know that that happens,

 

      and it happens to the pioneer industry, as well as

 

      the generic industry.  We need to get past those

 

      questions that really aren't meaningful and deal

 

      with really the scientific issues and look at what

 

      the risk is of some of these decisions that are

 

      made.

 

                MR. AHMED:  Exactly.

 

                                                               164

 

                DR. BOEHLERT:  So, no, I support what you

 

      are saying.

 

                MR. AHMED:  Thank you, because we know FDA

 

      has very limited resources.  We don't want to waste

 

      your time in terms of providing meaningless data.

 

                DR. KIBBE:  Thank you.

 

                MR. AHMED:  Any other questions?  Thank

 

      you.

 

                DR. KIBBE:  Gerry, you have the last word.

 

               Pharmaceutical Research and Manufacturers

 

                     of America (PhRMA) Perspective

 

                DR. MIGLIACCIO:  Three weeks ago, the FDA

 

      released their final report on the 21st Century

 

      Initiative.  This morning, Helen, Jon, and Ajaz

 

      talked about some gaps to achieving what was

 

      defined in that final report.

 

                That final report did a very good job of

 

      defining the desired state, and I am not sure how

 

      many of the committee members have gone through the

 

      entire report.  I had 12 hours on a flight to

 

      Tokyo, and I couldn't get through it all, but it

 

      does a very comprehensive job of painting the

 

                                                               165

 

      desired state.  Yes, there are gaps in the FDA and

 

      yes, there are gaps in industry.

 

                In the time that I have, I am not going to

 

      give PhRMA perspectives on every element of the

 

      final report. What I will do is touch on certain

 

      elements of it.

 

                I want to reinforce what Helen said

 

      earlier about culture change, make a couple of

 

      general comments on the report, and then talk about

 

      three of the documents in the final report, the

 

      Quality Systems guidance, the risk-based inspection

 

      model, and the ONDC risk-based quality assessment

 

      system, and then finally, summarize before lunch.

 

                Three years of culture change.  It's a

 

      two-year process, but we have been working on this

 

      for a lot longer than two years.  In fact, I

 

      contend that we have been working on this for three

 

      to four years, and the culture is changing.

 

                It is changing, it's gradual, in some

 

      cases it is dramatic and others, but there has been

 

      in this unprecedented period of communication and

 

      learning for both the industry and for FDA, and

 

                                                               166

 

      that has really been driven by the, first of all,

 

      having a shared vision of what the desired state

 

      is, maybe not always agreeing on how to get to the

 

      desired state, but at least having the shared

 

      vision of where we wanted to get to, and having

 

      open communications about that.

 

                PAT was the model, and much has followed

 

      based on the model.

 

                We, in industry, are moving from a fear of

 

      data to a passion for process understanding.  Now,

 

      let me describe that fear of data.  I grew up in

 

      this industry, I have been in it for 25 years, and

 

      I remember the days in the laboratory where I did

 

      some testing that was not required by the documents

 

      in my NDA, and getting reprimanded for doing any

 

      testing that was not required and where I did not

 

      have a clear understanding of how I was going to

 

      deal with the data.

 

                Well, we are getting much better now, we

 

      are using much more sophisticated tools at

 

      converting data into knowledge, and therefore, the

 

      fear of just data generation is going away, and we

 

                                                               167

 

      are also learning very rapidly that that data, if

 

      converted properly into process understanding,

 

      process knowledge, can tell us a lot about our

 

      processes, can tell us where the variability is,

 

      and can help us remove that variability where it is

 

      excessive.

 

                The next point, science, not blind

 

      compliance, is winning the day more often.  I am

 

      seeing this both internally and I am seeing it

 

      during inspections by FDA, that there is much more

 

      discussion.  There is much more discussion about

 

      the science, and I certainly, from my perspective

 

      in the industry, and I see my colleagues, as well,

 

      in other companies, they are using the science to

 

      make decisions much more frequently than simply

 

      sitting back and saying I am not going to take a

 

      risk, blind compliance, this is what the Agency

 

      expects, and just ignore the science.  So, that is

 

      going away.

 

                I think you are finding in many companies,

 

      innovation is accelerating, and a lot of it has to

 

      do with the open dialogue and the encouragement of

 

                                                               168

 

      the Agency obviously in this area.

 

                Interactions during inspections are

 

      changing, and I think some companies have seen

 

      dramatic changes, others subtle changes, but Helen

 

      talked I guess yesterday about the dispute

 

      resolution process, and I think that the

 

      discussions leading up to the guidance, the draft

 

      guidance, have had a significant impact on the way

 

      both our firms and the FDA investigators approach

 

      an investigation or an inspection.

 

                We are far more willing to put much more

 

      scientific knowledge on the table during an

 

      inspection.  Why?  Because basically, the dispute

 

      resolution process says that if you are going to

 

      dispute a scientific issue, you have to use what

 

      you have presented during the inspection, so we

 

      want to make sure we get that information out.

 

                Most of used to sit there and say, all

 

      right, we will wait to see if the investigator puts

 

      it on the 483, and if they put it on the 483, then,

 

      we will provide a written response to dispute it.

 

                Well, now, we are dealing with it during

 

                                                               169

 

      the inspection, and now the investigators are

 

      taking the time to look at the science, and they

 

      are calling in outside help when they need it to

 

      review the science, and we are seeing more and more

 

      issues resolved during the inspection, or shortly

 

      after the inspection with the district.

 

                So, I think the dialogue about dispute

 

      resolution has taken us 90 percent of the way to

 

      dealing with scientific disputes.

 

                So, some general comments on the final

 

      report.  I think this is a Churchill quote.  "This

 

      is not the end, it is the beginning."  The

 

      infrastructure is in place, but we have a lot to

 

      do.

 

                We applaud the magnitude of what has been

 

      accomplished, and not so much the volume of work

 

      that was accomplished by FDA with assistance from

 

      industry and academia, but the rigor.

 

                If you look at these documents carefully,

 

      it is not individuals sitting around and putting

 

      their own thoughts on paper, but bringing in

 

      experts from various fields, including risk

 

                                                               170

 

      management, and ensuring that the documents had the

 

      technical content that was required to achieve the

 

      objective.

 

                We obviously, in both industry and FDA,

 

      need to continue to work together to make sure that

 

      what is in those documents is fully understood

 

      throughout the Agency and throughout industry, and

 

      modified as appropriate, and then implemented.  So,

 

      we have a lot more to accomplish.

 

                Let me now turn to three of the documents

 

      that were in there.

 

                First of all, the Quality Systems

 

      guidance, and this is the Quality Systems guidance

 

      for industry.  We were very pleased to see the

 

      Quality Systems document for use within FDA.  We

 

      think that is going to contribute to their

 

      operation significantly, but the guidance for

 

      industry is very comprehensive, and we think it is

 

      of great utility.

 

                I know many companies are already using it

 

      to benchmark themselves, to look at our own Quality

 

      Systems and say how do we match up against this

 

                                                               171

 

      document.

 

                Now, there are some key issues in the

 

      document, and PhRMA will be commenting on this

 

      document and many others.  I am just trying to

 

      point out some high-level issues, but certainly

 

      process understanding, what level of process

 

      understanding leads to flexible continuous

 

      improvement in the regulatory environment?  It's a

 

      key issue and I think both Helen and Ajaz pointed

 

      that out this morning.

 

                We believe that it is imperative that

 

      these concepts be addressed on the global level.

 

      Somebody pointed out earlier today, most of us are

 

      in global businesses, the FDA guidance on Quality

 

      Systems, we believe really lays that starting point

 

      for ICH discussions on the proposed Q10.  So, we

 

      really believe that this needs to go on a global

 

      basis and can contribute globally.

 

                There are some parts of the guidance that

 

      we need to ensure we look at, potentially revise.

 

      We need to move away from some of the current

 

      compliance systems, and get us into a Quality

 

                                                               172

 

      Systems mode.  So, I will just give you one

 

      example.

 

                The guidance calls for the trending of

 

      data, encourages the trending of data as being part

 

      of a good Quality System structure.  The problem is

 

      some of the data we generate for compliance

 

      purposes provides no value on a trending exercise.

 

                How do you trend "none detected" or below

 

      level of quantitation?  How do you trend data that

 

      is rounded down to a point where it no longer has

 

      any meaningful benefit to gaining process

 

      understanding?  And why trend data that is

 

      generated that has no relationship to critical

 

      quality attributes or critical process parameters?

 

                Our current specification system drives

 

      the collection of that data.  So, we talked about

 

      specifications earlier.  I think this workshop in

 

      March is going to help us move these discussions

 

      forward, but our current specification system has

 

      to evolve.  It has to evolve so that we are

 

      collecting meaningful data on things that are

 

      critical to quality.

 

                                                               173

 

                Now, let me move to the risk-based

 

      inspection model, certainly a solid step forward to

 

      allow FDA to focus on higher risk sites.  We do

 

      question some of the elements of the algorithm, and

 

      we think they require further discussion.

 

                Again, as I raised at the Manufacturing

 

      Subcommittee meeting, and Judy talked about

 

      yesterday, the concept of volume is problematic to

 

      us, because if you have a manufacturing facility

 

      that makes two or three products at very high

 

      volume, actually, they understand those processes

 

      much more than a facility that may make 20 products

 

      at lower volumes, and they are probably much better

 

      controlled. Therefore, the risk associated with

 

      high volume is probably, in many cases, much lower.

 

                So, the use of a strict volume metric in

 

      the algorithm is problematic, and we would like to

 

      discuss that.

 

                But our most serious concern is about the

 

      transparency of the system.  These are words from

 

      the risk-based inspection model document, and what

 

      they say is that the FDA brought in experts, and

 

                                                               174

 

      the experts have established risk weightings for

 

      various processes, products, unit operations I

 

      assume, we don't have that information, and that

 

      has all been used to determine the risk factors or

 

      the risk level of various sites, but they don't

 

      intend to publish or disclose that information.

 

                Now, when we started this three years ago,

 

      we said one of the key objectives was that we

 

      understood where the risk was in this business, and

 

      that both industry and FDA focused on the high

 

      risk.

 

                Another document that was issued in the

 

      final report is this defining the customer in a

 

      regulatory agency, and we are, the industry is, a

 

      customer in certain case of the Agency, especially

 

      when it comes to their guidance documents and

 

      position papers.

 

                If you look in the middle, when FDA

 

      develops guidance documents representing the

 

      Agency's current thinking on a particular subject,

 

      we provide clarity and understanding to firms.

 

                Well, there is no clarity and

 

                                                               175

 

      understanding on what the FDA considers high risk

 

      in manufacturing coming out of this document.

 

      Also, in the final report, the FDA will now be

 

      using a Quality Systems approach to improve the

 

      predictability and consistency.  Again, there is no

 

      predictability here.

 

                We believe that industry and FDA have to

 

      have a mutual understanding of what processes, what

 

      unit operations are considered higher risk.  We

 

      believe experts in industry should have the

 

      opportunity to discuss and potentially debate what

 

      is considered higher risk by FDA, so that both

 

      industry and FDA can focus their resources on what

 

      we mutually agree are higher risk.

 

                We think transparency is essential here,

 

      and I truly believe this is a win-win-win, a win

 

      for FDA, a win for industry, and a win for the

 

      patients if we all agree and focus on what is high

 

      risk, but if it's not going to be published, if we

 

      are not going to understand whether the FDA

 

      perceives certain facilities as high risk, we are

 

      going to do our own risk assessment, and it may be

 

                                                               176

 

      different.  It may be different, and I don't think

 

      that is going to be an effective use of anyone's

 

      resources.

 

                So, we feel very strongly that

 

      transparency here is essential, and we hope FDA

 

      will continue the dialogue with us on this specific

 

      model.

 

                Finally, the ONDC risk-based assessment,

 

      quality assessment system, it represents a very

 

      profound change in the organization and review

 

      process.  You heard a lot about that both yesterday

 

      and today.  We support the objective strongly, but

 

      a lot of work is required to achieve the end state.

 

                Now, on the PAT Initiative and then on the

 

      GMP or the Drug Quality Initiative, the FDA

 

      demonstrated a willingness to put the leadership in

 

      place and the resources in place to make it happen,

 

      and I think this two-year report is an evidence of

 

      that.

 

                The question we have, a question that will

 

      we have the same commitment of leadership and of,

 

      in this case, review availability, to deal with

 

                                                               177

 

      this initiative, and as importantly, will industry

 

      have the same input into this process as we had

 

      into the PAT and the general Drug Quality

 

      Initiative over the past two to three years.  So,

 

      it is a key issue for us.

 

                There is this potential that this proposal

 

      creates this expectation.  I think, Judy, you said

 

      something about this yesterday in your summary of

 

      the Subcommittee meeting, that there is going to be

 

      a significant increase in the knowledge provided

 

      upfront.

 

                We will be providing different information

 

      and more knowledge, yes, not necessarily more

 

      information, it is going to be more knowledge.

 

                But again, there will be a greater degree

 

      of process understanding, but it still will be

 

      limited with the original NDA submission.  It will

 

      be sufficient to establish that we have a robust

 

      process, but process understanding is a continuum

 

      and will accelerate post-approval.

 

                So, you know, you are going to get the

 

      design space in the NDA, but that design space may

 

                                                               178

 

      be limited.  It will expand tremendously in the

 

      first one to two years of commercial manufacturing.

 

                We believe FDA should work through ICH to

 

      establish a global partnership for the risk-based

 

      quality assessment system.  Right now the

 

      assessment systems in Japan, the EU, and the U.S.

 

      are very different, and we certainly should not

 

      exacerbate the differences by moving forward with

 

      this initiative without including our global

 

      partners.

 

                One area which we understand the

 

      motivation to go to the pre- and post-marketing

 

      approach in the quality assessment system, but it

 

      does require some further evaluation.

 

                As I said just a few minutes ago, process

 

      understanding significantly increases during the

 

      first one to two years of marketing, and potential

 

      for continuous improvement is really the highest

 

      during that period of time, and there is certainly

 

      a value in having the reviewer who reviewed the

 

      original NDA involved in those initial continuous

 

      improvement changes.

 

                                                               179

 

                As I said, you have got a limited design

 

      space in the NDA, and that design space is going to

 

      grow exponentially in the first couple of years.

 

                Having to shift gears from one reviewer to

 

      another may be somewhat counterproductive.  So, we

 

      think this requires further discussion as to is

 

      there an appropriate time frame where the handoff

 

      goes to postmarketing.

 

                Helen talked this morning about two

 

      systems, potentially two review systems, one where

 

      you have the science process understanding in the

 

      application, one without.  The problem is that

 

      process understanding is not a yes or no answer,

 

      it's a continuum.

 

                So, every application, Company A may

 

      believe in this concept of manufacturing process

 

      and process understanding, but Company A's NDA for

 

      product X and for product Y will have different

 

      levels of process understanding.

 

                How will a reviewer determine where the

 

      application is in that continuum and how it should

 

      be addressed?  Key questions we think need to be

 

                                                               180

 

      addressed between FDA and industry as we move this

 

      quality assessment system forward.

 

                So, to summarize at two minutes before

 

      noon, Mr. Chairman, the infrastructure is in place,

 

      but some modification is required.  We are very

 

      pleased with the two-year report.  We think it is

 

      extremely comprehensive and really helps us focus

 

      on that desired state.

 

                The culture is changing, it needs more

 

      changing, but I think the steamroller is moving and

 

      I don't think it is going to stop either in

 

      industry or in FDA.

 

                The desired state is on the horizon, we

 

      are starting to see it in little bits at

 

      manufacturing sites and in research and development

 

      sites, you are starting to see the desired state

 

      coming to fruition, and I think it is not as far

 

      away as some might think.

 

                Thank you.

 

                DR. KIBBE:  Thank you.  I see by the clock

 

      on the wall that we are in plenty of time to

 

      actually get up and go to lunch.  We have no

 

                                                               181

 

      individuals who seek time during the open forum, so

 

      that we will return and at 1 o'clock we can

 

      entertain questions and comments about all of the

 

      speakers that we have heard prior to lunch break,

 

      and then we can jump right into the information and

 

      presentations in the second half of the day.

 

                I know that there are some members of the

 

      committee who need to get out of here by 3:30 or

 

      4:00 and we are going to try out best to make sure

 

      that the bulk of the work is done before they have

 

      to leave, and I appreciate your attendance.

 

                [Whereupon, at 12:00 noon, the proceedings

 

      were recessed, to be resumed at 1:00 p.m.]

 

                                                               182

 

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

 

                                                       [1:00 p.m.]

 

                DR. KIBBE:  I see by the clock on the wall

 

      that we should be turning on our equipment, so we

 

      can start recording the wonderful brilliance that

 

      is coming forth from these meetings.

 

                We agreed before we went to lunch that we

 

      would spend some time responding to this morning's

 

      activities and give an opportunity for those with

 

      extensive comments and questions to discuss the

 

      issues, and I know, Nozer, you are ready.

 

                Committee Discussion and Recommendations

 

                DR. SINGPURWALLA:  This is to Ajaz.  Would

 

      you like to sit down while I ask the questions, or

 

      would you like to stand?  You will stay here.

 

                DR. KIBBE:  It's safer, you are further

 

      away over there.

 

                DR. SINGPURWALLA:  Anyway, I enjoyed your

 

      presentation.  The ideas again are visionary and

 

      stimulating, and the comments I want to make are

 

      purely to improve upon what I think is something

 

      positive.

 

                                                               183

 

                The first question I want to ask you is a

 

      particular viewgraph that you put up in which you

 

      said the future definitions of quality should be

 

      probabilistic in nature.  This is a comment

 

      attributed to Janet Woodcock, although I am pretty

 

      sure you may have had the thought.

 

                What do you mean by that?  I subscribe to

 

      the view, but I would like to see what you want to

 

      say to it.

 

                DR. HUSSAIN:  I think, in part, that sort

 

      of follows a continuum of our discussions at FDA

 

      Science Board, and that is the IPAP RS debate also,

 

      is we often establish acceptance criteria, and so

 

      forth, in a more deterministic type of thinking in

 

      the sense of we don't utilize variability as a

 

      means of decisionmaking, and our criteria are 75 to

 

      125, that's it, and no unit outside that for a

 

      given sample, so it's an outgrowth of that thinking

 

      to saying that at one level, to say let's utilize

 

      the statistical distribution variability in making

 

      decisions.

 

                That is one level of that, but

 

                                                               184

 

      probabilistic also is linked to the risk-based

 

      aspect, and risk is probability for harm and

 

      consequences of that, and I think to bring that

 

      level of thought into how we approach quality.

 

                DR. SINGPURWALLA:  I think this requires

 

      much more thought and discussion, but I was curious

 

      as to what you had in mind.

 

                On your viewgraph 7, you have those

 

      3-dimensional illustrations of critical parameters,

 

      reliability, and life, shelf life is what you had

 

      in mind.  I could not get the essence of what it is

 

      that you were saying there, and the main reason I

 

      am raising this particular issue is next time you

 

      present it, you may want to rethink as to what it

 

      is that you want to communicate, and I didn't get

 

      the message.

 

                DR. HUSSAIN:  The message was in the sense

 

      I think when we often think about shelf life as a

 

      point estimate or as a endpoint determination

 

      perspective, and so forth, and what I liked about

 

      that viewgraph was it brings in the variability, it

 

      brings in an understanding of variability from that

 

                                                               185

 

      context and how variability changes.

 

                So, that was an illustration, and so it

 

      provides a means to think about reliability and

 

      shelf life together.  So, that was the

 

      attractiveness of that viewgraph.

 

                DR. SINGPURWALLA:  You mean the

 

      reliability of a drug?

 

                DR. HUSSAIN:  Product.

 

                DR. SINGPURWALLA:  Of a product.

 

                DR. HUSSAIN:  Yes.

 

                DR. SINGPURWALLA:  Well, when we normally

 

      talk about reliability of a product, it is the

 

      probability of it either living to some life length

 

      or doing it or performing satisfactorily or not, so

 

      you had a similar notion of reliability in mind?

 

                DR. HUSSAIN:  Yes.

 

                DR. SINGPURWALLA:  Let me go to your

 

      viewgraph No. 24.  I want to introduce a new

 

      concept based on that viewgraph.  This is on page

 

      12.  Perhaps I am going to introduce a new buzz

 

      word, which you may find attractive in the future.

 

      Are you ready?

 

                                                               186

 

                I think that buzz word may also apply to

 

      some of Gerry's presentation.  You are talking

 

      about substantial, moderate, and minimum as not

 

      being probabilistic.  You said something to that

 

      extent.

 

                DR. HUSSAIN:  We haven't defined the

 

      probability aspects to that.

 

                DR. SINGPURWALLA:  I want to introduce you

 

      to the following.  You perhaps know that in our

 

      presidential debates, President Bush used the word

 

      "fuzzy math," and I think now Kerry is talking

 

      about "fuzzy calculations."  But there is a body of

 

      knowledge called "fuzzy sets," and fuzzy sets are

 

      those whose boundaries are not sharply defined.

 

                So, the idea of substantial, moderate, and

 

      minimum would contribute to what is called a fuzzy

 

      set, whose boundaries are not sharply defined, and

 

      I think Gerry said something, I forget exactly

 

      where, but he was also alluding to a fuzzy set.

 

                Now, what I am recommending to the FDA and

 

      to the drug industry and whoever else is listening,

 

      that the notion of fuzzy sets may be very useful

 

                                                               187

 

      here, particularly with your nasal sprays

 

      thresholds, with the idea of a threshold. You

 

      cannot have a precise threshold.  You cannot say

 

      that if something is more than 4 inches, it is

 

      good, and if it is less than 4 inches, it is bad.

 

      The boundary is fuzzy.

 

                There are ways by which you can endow

 

      probabilities on fuzzy sets, and I would like to

 

      alert you to that particular body of knowledge, and

 

      I would like to alert you to looking into it as a

 

      possible venue for the kind of problems that you

 

      are working on.

 

                This is very recent, very new.  The idea

 

      of fuzzy sets has been around.  Control engineers

 

      use it, but to make it precise, and to endow

 

      probabilities on it is something very, very new,

 

      and the last issue of the Journal of the American

 

      Statistical Association has a paper with discussion

 

      on that particular topic.  I urge you to look at

 

      it.

 

                The last comment, and this is a comment,

 

      on your last slide, on immediate education needs. 

 

                                                               188

 

      First, I want to say what else you need into this

 

      package.  You really don't need introduction to

 

      statistical quality control.  What you need is

 

      introduction to statistical process control,

 

      because when you take quality control, it goes back

 

      to acceptance sampling, and you are really

 

      interested in a process.

 

                You say you should understand variability.

 

      I think the point is if you understand variability,

 

      variability goes away.  Once you understand it, you

 

      know what to do with it, and therefore it vanishes.

 

                What you really need as far as an

 

      educational component is some training and

 

      appreciation of what do we mean by uncertainty, and

 

      how do you work with uncertainty.  I think that is

 

      an important component.  Once that is understood,

 

      all other issues, process control, biostatistics,

 

      non-biostatistics, all those become very clear.

 

                The last question is how do you plan to

 

      implement this immediate education needs, what are

 

      you going to do, hold workshops?

 

                DR. HUSSAIN:  No, I think we have

 

                                                               189

 

      different mechanisms to sort of training.  We

 

      actually have formal training programs that

 

      actually go on, on a regular basis at a

 

      training--we used to have what I think we called

 

      staff college.  There are aspects of that.  There

 

      is an Office of Training and Communication, which

 

      is thinking of a course on design of experiments,

 

      and a whole series of courses which are sort of

 

      built in.  People can take them whenever they have

 

      time, so these are ongoing.

 

                But immediately what we have been focused

 

      on is workshops, internal seminar series, and so

 

      forth.  We are starting in a step-by-step fashion.

 

      As we move towards the final guidances, for

 

      example, we generally have a training around the

 

      guidance.  Much of that we also get captured there.

 

                But my thoughts are I think we need a

 

      portfolio of courses that are available every

 

      quarter or every semester, or whatever, that people

 

      can take on a regular basis, so we need a

 

      curriculum that is available for our staff whenever

 

      they choose to take it, and so forth.

 

                                                               190

 

                DR. SINGPURWALLA:  My reaction to these

 

      things, available on a semester-by-semester or

 

      quarter-by-quarter is that they fail.  What you may

 

      want to do is have special courses like this

 

      throughout the FDA, not just your organization,

 

      provided they agree that this is what is needed,

 

      and have them, you know, once, and maybe twice, and

 

      then stop, but then require that the people who

 

      should need that exposure go.

 

                If you keep it optional, somebody says,

 

      well, I know statistics, I know what r-square is, I

 

      don't need to take a course.  That is where the

 

      tragedy is, that everybody thinks they know it.

 

      So, I propose that you design something specific

 

      and go through that.

 

                Again, I am not volunteering since I made

 

      the suggestion.

 

                DR. KIBBE:  Do you have a suggestion for a

 

      college that they could take all these courses at?

 

                DR. MORRIS:  Someplace in Indiana maybe,

 

      Notre Dame.

 

                DR. KIBBE:  I was hoping for Pennsylvania

 

                                                               191

 

      myself.

 

                DR. MORRIS:  Pennsylvania, okay.

 

      Actually, the University of Hawaii I think would be

 

      good.

 

                I actually had a couple of comments I

 

      think that go across Helen's, Ajaz's, and Jon's

 

      talk, but I want to preface it by saying that in

 

      the time I have been spending with you all, it

 

      occurs to me that there is a lot that is right,

 

      right now, with the reviewers and the reviewing

 

      process that we don't want to lose.  I don't get

 

      paid any extra for that, by the way.

 

                In that sense, the idea of a two-tiered

 

      approach doesn't really seem to have legs to me.

 

      In a sense what we are saying is, is that the

 

      reviewers would have to be somehow two tiered, and

 

      that is really not the way they review.

 

                What we talked about at the last

 

      Manufacturing Subcommittee was the idea that when

 

      companies can provide a rationale with supporting

 

      data, that that automatically registers as a level

 

      of understanding as opposed to when the reviewers

 

                                                               192

 

      have to develop a rationale based on the data that

 

      they have available, but at the end of the day,

 

      they are still looking for rationales, still

 

      looking for flags in terms of safety and efficacy

 

      and rationales.

 

                So, I don't really see that there is a

 

      real need for that.  I mean we can certainly

 

      comment on it, and probably should hear from Paul

 

      and Gerry on it.

 

                I have a couple of other comments, but go

 

      ahead.

 

                DR. HUSSAIN:  No, I think that has been a

 

      concern, and this was an extensive topic that we

 

      discussed at the London meeting of ICH, especially

 

      working group meeting on that.  The language we

 

      have crafted, I mean that took a long time to

 

      craft, to say there won't be any two systems or not

 

      tiers at all, in the sense it is a smooth

 

      transition.

 

                But the reality is you will have much more

 

      varied types of submissions.

 

                DR. MORRIS:  Absolutely.

 

                                                               193

 

                DR. HUSSAIN:  But that is managing that,

 

      and so forth.

 

                DR. MORRIS:  And I agree, but I think in

 

      that sense, you know, there were a couple of

 

      things.  You know, the evaluation of the

 

      Pharmaceutical Development Report that you had

 

      mentioned, Helen, and really it should create less

 

      questions if you assume that most questions come

 

      from the lack of a presentation of a rationale, so

 

      hopefully, we will realize some efficiencies from

 

      that.

 

                Similarly, Ajaz, I think the review of the

 

      critical variables and attributes should be

 

      improved by the Development Report, the P2 and the

 

      CTD, as well.

 

                I guess this point you had made in your

 

      presentation, as well, I can't remember if it was

 

      Jon or you, Ajaz, but the idea that there are

 

      already in industry, the scientific expertise.  It

 

      is a question of how they are organized and linked

 

      in terms of communication.

 

                I think the same is true in the Agency. 

 

                                                               194

 

      There is a good bit that is already in place, that

 

      just needs to be wired, and that is really all I

 

      had to say.

 

                DR. KIBBE:  Melvin, did you have

 

      something?

 

                DR. KOCH:  I had a comment or question for

 

      Jon.  It had to do with the new process or actually

 

      even the old process in terms of creating, say, a

 

      draft or a guidance, and after you go through the

 

      legal audit, the public comment, and eventually

 

      have a final draft, I am just wondering if there

 

      isn't--maybe I should ask the question--is there

 

      anything that happens before the final draft in

 

      terms of explaining how the comments were used?

 

                I know they all show up on a docket, but I

 

      know with the most recent PAT final guidance, there

 

      were questions that those who are hoping to use it

 

      have, mainly because there is some part of it which

 

      seems to refer back to doing things the way they

 

      used to be, and there is a little bit of inhibition

 

      initially in how to use it.

 

                I am just wondering, in the process, you

 

                                                               195

 

      mentioned something called a public draft, and if,

 

      say, after the public comments are taken, you know,

 

      the almost final draft shows up for some quick

 

      response.

 

                MR. CLARK:  The comment that we receive

 

      when--this is true of the old process and true in

 

      new, as well--public comments received are treated

 

      as if they are FOIable whether they are or not.

 

      So, there is a documentation of comments, an

 

      indication of how they are collated, and the

 

      response to them if they are grouped together as a

 

      group or individually depending, that is up to the

 

      group whether they group them together or not.

 

                The FOI rules, they shift a little on me,

 

      I am not sure what level they are available, but we

 

      treat them as though they are.  It's a public

 

      draft, it was not meant to be any different than

 

      the current draft that we now publish. We provide a

 

      Notice of Availability in the Federal Register and

 

      then it is provided on the web.  That draft is what

 

      receives the public comments.

 

                What I was referring to, the main crux of

 

                                                               196

 

      what I wanted to have accomplished here was to

 

      involve this body in evaluation of prioritization

 

      of guidances in general and where we would need

 

      them.

 

                In other areas where we talked about this,

 

      it had come up that we needed to have a better way

 

      of looking at whether or not we needed a guidance

 

      in certain instances, because our methods for doing

 

      that now usually involve public workshops, very

 

      driven by the FDA, and perhaps we should include

 

      some more outside opinion as to where we would need

 

      guidance.

 

                DR. KOCH:  I guess I was wondering is

 

      there a process for revision and/or interpretation

 

      of a final guidance?

 

                DR. HUSSAIN:  No, I think the policy in a

 

      sense, we do not plan to sort of outline how we

 

      address the comments, only for regulation changes

 

      that is a requirement.  For guidances, comments,

 

      essentially, we don't share that information.

 

                MR. CLARK:  If we receive some indication

 

      that there are interpretations of the guidance that

 

                                                               197

 

      aren't going the way we anticipated, this is a

 

      point of having a question and answer associated

 

      with that document or with that topic.

 

                DR. KOCH: Right, and it is in that

 

      context.  Anyway, we maybe could talk at some point

 

      in terms of some specifics.

 

                DR. KIBBE:  Pat.

 

                DR. DeLUCA:  I just want to comment, and I

 

      guess I best comment using Ajaz's slide, the cGMP

 

      Initiative.  It is on page 14.  It has been my

 

      experience that most times in the pre-IND, pre-AND

 

      product development process development, that these

 

      is a hesitance at actually getting too innovative

 

      or pursuing new things and trying to improve a

 

      product with the idea that, oh, well, after it is

 

      marketed, then, we will do this, which I don't

 

      think in my experience that it happens very often.

 

                So, I like this.  It is because you have

 

      in the circle here that after you get to improve

 

      it, and there is a slot there for continuous

 

      improvement and innovation, and I think I may have

 

      said this before at previous meetings that I think

 

                                                               198

 

      even after postmarketing, that there should be

 

      vigilance in trying to improve the process, and

 

      sometimes you have to try to change both in order

 

      to do one or the other.

 

                I am certain, seeing here that the

 

      expertise certainly exists in the industry the

 

      expertise exists here, and kind of encourage that

 

      it looks like the mentality may even be moving in

 

      that direction, too.

 

                So, that is something that I would

 

      encourage as the desired state, that postmarketing,

 

      that there is this effort, continued effort to

 

      improve the product and the process.

 

                DR. KIBBE:  Gordon, you have a comment.

 

                DR. AMIDON:  Yes, I have a comment on two

 

      of the slides that Ajaz presented.  I mentioned one

 

      to you at lunch, Ajaz.  The other was--

 

                DR. KIBBE:  Just give a page number.

 

                DR. AMIDON:  On page 11 in your

 

      presentation, the top slide.  Interpretation and

 

      Optimization of the Dissolution Specifications for

 

      a Modified Release Product with an In Vivo-In Vitro

 

                                                               199

 

      Correlation.

 

                I am not sure what implication the authors

 

      were making with the dissolution passed, the

 

      bioequivalence passed and the dissolution passed,

 

      bioequivalence failed, but my question would be

 

      what dissolution did they do.

 

                The impression I had from your

 

      presentation was it was suggesting that dissolution

 

      was inadequate in predicting bioequivalence, and it

 

      obviously wasn't, but that is because they did the

 

      wrong test, the dissolution test.

 

                So, this implies to me that we need to

 

      really be more specific when we are talking about

 

      dissolution, what is the test, did the test reflect

 

      what is going on in vivo, because if your in vitro

 

      test reflects your in vivo process, that has to

 

      predict what is going on.  Otherwise, there is

 

      magic in between.

 

                DR. HUSSAIN:  I think the theme of that

 

      paper was twofold in the sense to evaluate--if you

 

      have the handout, it is part of that--the team was

 

      to evaluate the linkage between in vitro

 

                                                               200

 

      dissolution and in vivo bioequivalence and how

 

      variability or random variability sort of plays

 

      into that.

 

                Authors looked at what they call a

 

      non-optimal specification and an optimal

 

      specification, optimized specification based on in

 

      vitro correlation, then essentially simulated with

 

      random variability built in, in vitro, as well as

 

      in vivo, what is the likelihood of finding failed

 

      dissolution, so that has been that analysis.

 

                DR. AMIDON:  I didn't mean for you to have

 

      to defend the paper, and I will have to take a look

 

      at it, but there is something wrong here in my mind

 

      at least based on this one slide, because if your

 

      in vitro dissolution reflects the in vivo

 

      dissolution process, it will predict

 

      bioequivalence.

 

                DR. HUSSAIN:  No, that is the whole point

 

      here in a sense.  The point here, all we can do

 

      with the current dissolution methodologies are the

 

      mean values.  We have no idea on the variability.

 

      So, once you factor in the variability, then, you

 

                                                               201

 

      see these aspects.  That is the crux of that.

 

                DR. AMIDON:  The second comment would be

 

      on just the previous page, page 10, at the bottom

 

      of the slide, which I did mention to you at lunch.

 

      I am confused whether this is a flow chart or an

 

      event chart of something.

 

                The very first step, does dissolution

 

      significantly affect bioavailability, I mean the

 

      answer, I have problem with the answer "No,"

 

      because I can make a tablet no matter what the drug

 

      properties, it will not dissolve and disintegrate,

 

      so dissolution always affect bioavailability, it is

 

      a matter of how much.

 

                I guess my broad comment, and I will make

 

      this again later in my presentation, is that we

 

      need more research on dissolution particularly in

 

      vitro to in vivo. That is where we have the big gap

 

      in our scientific understanding today.

 

                DR. HUSSAIN:  I think this is an ICH Q6A

 

      decision tree, and now looking back after years of

 

      this, I do want to give credit to Professor Nozer

 

      Singpurwalla that he really pointed out this is an

 

                                                               202

 

      event tree, not a decision tree, and I actually

 

      before that had not paid attention to that fact.

 

                Now, when we look at it after years, some

 

      of the questions really don't make sense.

 

                DR. KIBBE:  Michael.

 

                DR. KORCZNSKI:  This is just a comment

 

      related to the past two days of discussion.  I

 

      think we really heard some excellent data being

 

      generated from the FDA labs, and I might add, after

 

      spending a number of years in the industry, I wish

 

      some of my retired colleagues could have heard the

 

      discussion.  I think they would have marveled at

 

      the progress being made scientifically.

 

                A thought comes to mind.  I think this is

 

      a very significant question.  I have tried to

 

      distill a significant thought here.  Can innovative

 

      drugs, as a result of where we are going and the

 

      data we are seeing, eventually be reduced in scope

 

      relative to clinical trials and without a loss of

 

      patient safety or efficacy?

 

                Just to kind of encapsulate that, if,

 

      indeed, you do have some very excellent innovative

 

                                                               203

 

      types of assays at the preclinical level, such that

 

      they perhaps might even mitigate to some degree or

 

      reduce Phase I studies, because they are so

 

      thorough and better predictors, and then if you got

 

      to that stage, could there be a consideration of

 

      collapsing Phase I and Phase II clinicals into one

 

      clinical trial basically, then, with all the data

 

      that we saw from the ICSAS staff, the computerized

 

      data, is it possible in some way to network with

 

      sponsor companies, such that more drugs could

 

      probably be placed in an orphan drug clinical

 

      pathway as opposed to the current conventional

 

      Phase III.

 

                So, long term, there are some real

 

      opportunities, I think, in condensing and

 

      collapsing that costly and lengthy clinical trial

 

      procedure, and the data to a large extent that is

 

      being collected.

 

                Just another two items.  One, we talked

 

      about innovative drugs.  I think what is going to

 

      be important, this is kind of futuristic, would be

 

      the identification of target sites for disease

 

                                                               204

 

      states, and very important with the development of

 

      these nanomolecules and small peptides will be the

 

      delivery systems.  What will be the micro delivery

 

      system to the host site especially as we talk about

 

      gene therapy?

 

                So, the development of that technology has

 

      to occur in concert with the development of the

 

      molecules, so we are going to have the molecules

 

      and not the means to deliver to disease state host

 

      sites.

 

                So, that is just an encapsulation.  Thank

 

      you.

 

                DR. HUSSAIN:  On the last point, I think

 

      there has been an amazing growth in that particular

 

      area, vehicles, nanovehicles, nanodrug delivery

 

      systems, and so forth, and we hope to publish a

 

      paper soon on the topic of dendrimers and then how

 

      dendrimers can sort of shrink promers [ph], and so

 

      forth, so there has been wonderful progress even on

 

      that side, on the delivery part of it.

 

                DR. KIBBE:  Anybody else?  Okay.  So, we

 

      kind of beat that into the ground, and we got that

 

                                                               205

 

      under control.

 

                We need to move forward.  I see that

 

      Lawrence is getting ready to go.  He has two sets

 

      of presentations, and he agreed that if we would

 

      just do exactly what he says in the first set of

 

      presentations, he would be really fast on the

 

      second set.

 

             Pharmaceutical Equivalence and Bioequivalence

 

                            of Generic Drugs

 

              The Concept and Criteria of BioINequivalence

 

                      Concept of BioINequivalence

 

                DR. YU:  Good afternoon.  This afternoon

 

      we have to deal with two topics.  One is follow-up

 

      topics which we have presented this committee six

 

      months ago, and a second topic, introduction topic,

 

      is called Bioequivalence of Locally Acting GI

 

      Drugs.

 

                Let me go through the first topics of

 

      bioINequivalence, concept and definition, which

 

      this topic, as I said, was presented to you six

 

      months ago on exactly April 14, 2004.

 

                The bioavailability is defined as the rate

 

                                                               206

 

      and extent of drug absorption.  Regularly, we use

 

      the Cmax as a surrogate for the rate absorption and

 

      the AUC, all to the exact area under the curve used

 

      for the extent of the drug absorption.

 

                So, the bioequivalence is defined as

 

      absence of a significant difference in the rate and

 

      extent absorption, and many other things, for

 

      example, become available at the site of drug

 

      action when administered at the same molar dose

 

      under similar conditions in an appropriately

 

      designed study. So, this is basically the CFR

 

      definition.

 

                With bioequivalence, we usually use 90

 

      percent confidence interval for AUC or the extent

 

      of absorption, or Cmax rate of absorption between

 

      80 and 125 percent.

 

                The passing bioequivalence criteria or

 

      confidence interval allows market access.

 

      Certainly, we have to, for example, generic firm

 

      have to meet other quality standards.  If you pass

 

      the confidence interval, or we call the

 

      bioequivalence for generics, this means the generic

 

                                                               207

 

      approvals, for innovators, this means demonstrated

 

      to be marketed formulation to be equivalent to

 

      clinical formulation.

 

                Those concepts are well understood, well

 

      developed, widely used.

 

                The question remains why do we need to

 

      define bioINequivalence?  It is because FDA

 

      receives studies, as we discussed six months ago,

 

      receives studies that attempt to reverse a previous

 

      finding of bioequivalence, in other words,

 

      companies, whether innovator companies or generic

 

      companies out there, to conduct a study, say, the

 

      generic products or products on the market, in

 

      fact, is a bioINequivalent.

 

                The bioINequivalent definition is not very

 

      well defined.  In many cases, to be scientific

 

      term, those studies actually should be defined as

 

      failed to demonstrate bioequivalence to be exact.

 

      That is part of reason when you define the criteria

 

      of a bioINequivalence, when you define the concept

 

      and criteria of bioINequivalence.

 

                The question certainly, what should

 

                                                               208

 

      bioINequivalence mean?  Bioequivalence leads to

 

      market access, bioINequivalence leads to market

 

      exclusion.  That is what

 

      bioequivalence/bioINequivalence basically means.

 

                Now, come back to say look at the results,

 

      what does it specifically mean when I said failed

 

      to demonstrate bioequivalence, failed to

 

      demonstrate bioINequivalence, bioequivalence or

 

      bioINequivalence, those concepts.

 

                In the center, the top one is demonstrate

 

      bioequivalence just because the confidence interval

 

      for this study is within the bioequivalence

 

      interval of 80 to 125 percent.

 

                The bottom one is demonstrated

 

      bioINequivalence, I use BIE here, it stands for

 

      bioINequivalence.  Now, the middle one, basically,

 

      the left side or right side, we have fail to

 

      demonstrate bioINequivalence and fail to

 

      demonstrate bioequivalence.  This basically is

 

      because neither, whether either outside of the

 

      confidence interval, outside of bioequivalence

 

      interval, 80 to 125 percent or not completely

 

                                                               209

 

      inside.

 

                Now, since the April discussion, I

 

      received several phone calls and also we had a

 

      discussion within FDA with my colleagues, some

 

      question came back that by definition of

 

      bioINequivalence may be too far.  Some people

 

      suggest maybe use mean of the point estimate,

 

      confidence interval, while the suggestion was that

 

      if you use the point estimate or mean ratio outside

 

      or above 125 percent, it should be defined as

 

      bioINequivalence.

 

                Now, certainly this makes a lot of sense.

 

      However, if we look at it deeper, statistically, it

 

      is not.  Let's look at an example here.  The top

 

      one, obviously, it failed to demonstrate

 

      bioINequivalence.  Now, failed to demonstrate

 

      bioINequivalence could have been for many reasons.

 

      One of the reasons you would think is not enough

 

      subjects, for example theoretically, they should

 

      have used 100 subjects or 50 subjects, and this

 

      study only used 10 subjects.

 

                Because of the small number of subjects,

 

                                                               210

 

      it makes the confidence interval of the final

 

      results much wider, therefore, end up you have a

 

      failed to demonstrate bioINequivalence.  We see we

 

      cannot use this result, failed to demonstrate

 

      bioINequivalence as definitive answer, is because

 

      if you use large number of subjects, there is two

 

      possibilities as you can see here.

 

                Could be bioINequivalence completely below

 

      the 80, outside of the bioequivalence confidence,

 

      either below 80 or above 125 percent.

 

                There is another possibility that even

 

      though the study, this is a small number of

 

      subjects study shows bioINequivalence, in reality,

 

      they could be bioequivalent, as you show here on

 

      the left side, right side or left side. So, the

 

      right side one is because once you use large

 

      numbers of subjects, could give you a definitive

 

      answer, you end up even though the study itself

 

      fails to show bioINequivalence, but at the end you

 

      have two outcomes, and that is the power that could

 

      be demonstrated bioINequivalence or demonstrated

 

      bioequivalence.

 

                                                               211

 

                That is part of the reason we say we may

 

      not be able to use these results to have a

 

      definitive answer to make regulatory decisions.

 

                The objective is the same which we were

 

      discussing back to April 14th, to develop a

 

      bioINequivalence criteria that are scientifically

 

      sound, statistically valid, fair to all parties,

 

      and easy to use.

 

                I want to remind you that the comments or

 

      conclusion draws back to the last discussion.  The

 

      first question, back to April 14th:  Does the ACPS

 

      agree with the distinction between demonstrating

 

      bioINequivalence and failure to demonstrate

 

      bioequivalence?

 

                The answer was yes.  That was the

 

      conclusion reached back to April 14, 2004.

 

                The second question:  Does the ACPS

 

      recommend a preferred method for evaluating the

 

      three pharmacokinetic endpoints for

 

      bioINequivalence?

 

                There are many sub-questions.  Here are

 

      the comments which were discussed.

 

                                                               212

 

                The committee agreed on a general

 

      understanding of bioINequivalence to move forward

 

      by recognizing this is not a simple matter.  In

 

      addition, the members felt that there is an

 

      important concept, especially now it applies to

 

      entire regulatory scenario.  There was no consensus

 

      at this point as to a final criteria pertaining to

 

      the three pharmacokinetic endpoints.

 

                We will present you today our

 

      recommendations based on those discussions and hope

 

      we can follow the comments or discussion.

 

                DR. MEYER:  Could we ask a question before

 

      we get confused with statistics?

 

                DR. KIBBE:  Okay, great.  Why don't we ask

 

      the question before we get statistical.

 

                DR. MEYER:  I like to approach things in a

 

      very simple manner.  If you could get slide 7 back,

 

      Lawrence.

 

                It strikes me that you are trying to use

 

      phraseology to fit a subsequent statistical

 

      analysis, and I am wondering if there isn't a

 

      simpler way to go about it.  I am kind of, of the

 

                                                               213

 

      school that you are either pregnant or you are not,

 

      and if you haven't taken the test yet, you can't

 

      say.  So, that is basically where I am going to go.

 

                I am going to number these 1, 2, 3, 4, 5.

 

      I think we can all agree that No. 1 is

 

      bioequivalent, and No. 4 and 5 are bioINequivalent,

 

      just as you have shown.

 

                DR. YU:  Yes.

 

                DR. MEYER:  I say No. 2 is

 

      bioINequivalent, not failed to demonstrate

 

      bioequivalence, the pregnancy/non-pregnancy thing.

 

      I say that because the mean is well outside 80

 

      percent, and increasing the numbers may shift the

 

      mean one way or the other.  You shifted it, of

 

      course, to the left to make it worse.

 

                I would say maybe it will stay the same.

 

      All I know is that the means are terrible, and the

 

      confidence limit kind of extends over to acceptable

 

      range, but in my view, if somebody came to me with

 

      that data and said I ran the study on 40 subjects,

 

      should I do 80, I would say no, reformulate.

 

                If they came to me with a confidence level

 

                                                               214

 

      that lopped that over, I would say, yeah, do an

 

      added number.

 

                So, I think the N problem, which could

 

      expand confidence limits, can be solved by

 

      requiring anyone that dose a bioINequivalence study

 

      to fix their N at the same as the person that got

 

      the approved ANDA.  So, if it took Teva 485

 

      subjects to do their study, then, I think Pfizer

 

      ought to do 485 subjects if they are going to try

 

      to prove that Teva is no longer bioequivalent for

 

      some reason.

 

                No. 3, I would say that fails to

 

      demonstrate bioequivalence by the current

 

      standards, but it also fails to demonstrate

 

      bioINequivalence, because the means are well below

 

      125.  That is a wash.

 

                You can't tell one way or the other,

 

      therefore, whoever is doing the study can't make a

 

      claim of bioINequivalence, because there is

 

      ambiguity in the data.  That is a much simpler

 

      approach than trying to come up with a new metric

 

      of 3 parameters or 1 parameter or what have you.

 

                                                               215

 

                DR YU:  Indeed, Marvin, you have excellent

 

      questions.  That is true, I guess, we have too many

 

      discussions on this topic.

 

                Well, come back to your question, is that

 

      does fail to demonstrate bioINequivalence, top, No.

 

      2, is actually demonstrated bioINequivalence here,

 

      that is the question.

 

                Indeed, you point out it probably could be

 

      bioINequivalence if the sponsors, whoever conduct

 

      the study, you have sufficient power, was

 

      sufficient in number of subjects.

 

                I guess the question comes back under this

 

      scenario, that for top one, which is clear,

 

      demonstrate bioequivalence, and the bottom one is

 

      clear, demonstrate bioINequivalence, however, for

 

      No. 2 or No. 3, this does not

 

      necessarily--especially for No. 2--if we receive

 

      the data, this does not necessarily suggest, we are

 

      not going to take any action whether or not you

 

      look at it, simply because you fail to show

 

      bioINequivalence, therefore, we are not going to

 

      take a look at it, we are not going to take any

 

                                                               216

 

      action, that is not the case.

 

                Certainly, this case, if you submit it to

 

      the Agency, and the study is well conducted, well

 

      powered, we have to look at all of the scenario and

 

      then to draw a scientific decision, or I guess what

 

      I am saying is top 1, and top 4 and top 5 give us

 

      definitive answer, top 2 and top 3, we have to look

 

      at it case by case.  We cannot draw very decisive

 

      conclusive decisions is part of the reason you have

 

      assumption here, you have a sufficient number of

 

      subjects.

 

                If you don't use a sufficient number of

 

      subjects, say, you only use 1 or 2 subjects,

 

      certainly, we were not able to say you have

 

      demonstrated.

 

                I hope I answered your question.  I guess

 

      we answer your "if" questions.

 

                DR. MEYER:  How about fixing the N at the

 

      same as the ANDA submission used?

 

                DR. YU:  That is one of the options, yes.

 

                DR. KIBBE:  You are responding to a

 

      question that I don't think the Agency is asking. 

 

                                                               217

 

      You are responding to a question that a sponsor of

 

      one of those studies would ask in order to get the

 

      study to do what it wants to do.

 

                Then, you are saying, well, the Agency

 

      should say if they did this, then, they probably

 

      would show this and what should we do about that.

 

      The difficulty for me in this whole scenario is

 

      that the failure to demonstrate bioequivalency

 

      doesn't necessarily prove bioINequivalency, and if

 

      a product is already on the market because it has a

 

      bioequivalency approval, what level of information

 

      do we need to reverse that decision.

 

                I agree with Lawrence, those two in the

 

      middle wouldn't justify in my mind as a regulator a

 

      change in the previous decision, whatever it was.

 

      Okay?

 

                DR. MEYER:  If the orange one, No. 3, was

 

      done under the same conditions as the ANDA holder.

 

                DR. KIBBE:  If it was done under the same

 

      conditions as the ANDA holder, I wouldn't reverse

 

      my decision on anything.

 

                DR. MEYER:  That is an exaggeration

 

                                                               218

 

      perhaps because let's say it's 127, you can't get

 

      real excited about that.  No, it would have to be

 

      125, but let's say it was 145.

 

                DR. KIBBE:  What I am saying is we already

 

      have a product that passed once.

 

                DR. MEYER:  Right.

 

                DR. KIBBE:  That study doesn't help me

 

      decide to reverse that decision.

 

                DR. MEYER:  The product has changed.

 

      Remember, Gary Levy published The Clay Feet of

 

      Bioavailability or Bioequivalence Testing.  You do

 

      it once on a hand-picked lot against one lot of the

 

      innovator product, a fresh lot of yours, and one

 

      you have selected maybe out of 12 of theirs, and

 

      then somebody else comes along with an older lot of

 

      theirs or vice versa, and no longer are you

 

      bioequivalent.

 

                DR. KIBBE:  I understand the argument.  I

 

      am just saying that, as a regulator, I wouldn't

 

      change anything I have got on the books based on 1,

 

      2, 3.  Okay?

 

                DR. YU:  I guess I will make one more

 

                                                               219

 

      comment. When you conduct bioequivalence studies,

 

      you look at availability, you look at the power,

 

      you look at the subject.  You are basically always

 

      saying here that the bioequivalence criteria is 80

 

      to 1 to 25 percent.  Agency never defines how many

 

      subjects you use.  You could have used 24, 48, 96,

 

      500, for example, for clinical endpoint studies.

 

                So, when you define the number of

 

      subjects, then the confidence interval could be

 

      variable, one way or another.  You define one, and

 

      you have another criteria.

 

                I think that for the bioequivalence

 

      criteria, we define the confidence interval instead

 

      of define the number of subjects.

 

                DR. KIBBE:  Go ahead, Nozer.  We are

 

      having a lot of fun here.

 

                DR. SINGPURWALLA:  I think it is always

 

      fun when committee members disagree, but something

 

      bothers me about this whole concept.

 

                DR. YU:  That was not the--

 

                DR. SINGPURWALLA:  That was not the

 

      question, I understand.

 

                                                               220

 

                DR. YU:  That was not the question.

 

                DR. SINGPURWALLA:  I understand, that was

 

      not the question.

 

                DR. YU:  If we don't want to live with it,

 

      I guess the decision will have to be made.

 

                DR. SINGPURWALLA:  I am sorry, my comment

 

      here is you are building a castle on sand, I think

 

      this whole idea--

 

                DR. KIBBE:  Or clay, right?

 

                DR. SINGPURWALLA:  Whatever you want to

 

      use, it's a castle that cannot hold up.  What you

 

      have done is you looked at Cmax and you looked at

 

      AUC, and if the Cmax and AUC are not significantly

 

      different, you say there is bioequivalence.  Not

 

      true?  What is it then?

 

                DR. KIBBE:  If you are going to give an

 

      answer, Don, you might as well get on a microphone.

 

                MR. SHERMAN:  Lawrence showed a quote from

 

      the regulations on the definition of

 

      bioequivalence, and it said rate do not show a

 

      significant difference.  The word "significant" in

 

      that sentence does not mean statistically

 

                                                               221

 

      significant, it means significant the way the word

 

      is used in the English language, substantial,

 

      important.

 

                DR. SINGPURWALLA:  Then, my comment

 

      becomes even more acute.  The whole thing should be

 

      relooked, revisited, because I kind of agree with

 

      Lawrence about those two, and I agree with our

 

      chairman about the two middle ones as demonstrating

 

      failure to demonstrate, then demonstrating. I mean

 

      I wouldn't change anything, but I think the whole

 

      concept of doing all this through this particular

 

      vehicle of setting confidence limits and looking at

 

      the little tail falls here or there seems

 

      completely capricious to me, and you may want to

 

      revisit this whole topic.

 

                DR. KIBBE:  We have been revisiting this

 

      since 1970 at least.

 

                DR. SINGPURWALLA:  I was not there.

 

      Change the paradigm.

 

                DR. KIBBE:  We are not revisiting the

 

      paradigm.

 

                MR. SHERMAN:  I just want to correct the

 

                                                               222

 

      notion that you look at Cmax and AUC and you

 

      approve products as inequivalent if there is not

 

      statistically significantly difference.  That

 

      hasn't been true for decades.

 

                DR. SINGPURWALLA:  So, it has been

 

      nonsense for decades.

 

                MR. SHERMAN:  You are entitled to your

 

      opinion, sir.

 

                DR. YU:  We will move on to the next

 

      topic.  Thank you.

 

                      Criteria of BioINequivalence

 

                DR. LI:  Good afternoon.  My name is Qian

 

      Li.  I am from Office of Biostatistics in CDER,

 

      FDA.  I am going to present a statistical criteria

 

      for evaluating bioINequivalence using multiple

 

      endpoints. I know there have been citing for using

 

      one endpoint, but I decided to move on to talk

 

      about multiple endpoints.

 

                Before I start to talk about the criteria,

 

      I would like first to discuss the question why we

 

      need to use multiple PK endpoints to assess

 

      bioINequivalence.

 

                                                               223

 

                To answer this question, we need to

 

      understand that for systematically delivered drug

 

      product, bioequivalence established by comparing

 

      the rate and extent of drug absorptions.  The rate

 

      and extent are usually represented by Cmax, AUCt,

 

      and AUCinfinity.

 

                In statistical terms, Cmax, AUCt and

 

      AUCinfinity, I refer to as PK endpoints.  For

 

      bioequivalence assessment in generic drug approval,

 

      it has evolved to use AUCt, AUCinfinity, and Cmax.

 

      As Lawrence has mentioned before, that you have to

 

      prove that 0 to 3 PK endpoints to be equivalent in

 

      order for the generic drug product to have market

 

      access.

 

                Now, for bioINequivalence, it can be

 

      established if one of the PK endpoints are

 

      inequivalent in truth.

 

                Now, in reality, we do not know the truth,

 

      so we have to perform statistical analysis to test

 

      all the PK endpoints.  That is why we need to

 

      assess multiple PK endpoints.  I hope this is clear

 

      to everybody now.

 

                                                               224

 

                Now, this is the outline of my

 

      presentation.  I will give a brief review on the

 

      criteria for bioINequivalence using one PK endpoint

 

      and then present strategies for assessing

 

      bioINequivalence using three PK endpoints, and I

 

      will discuss available approaches and then present

 

      power comparisons of those approaches.

 

                Then, I will discuss FDA's recommendation

 

      of using the three PK endpoints in assessing

 

      bioINequivalence.

 

                Now, let's first look at definition of

 

      bioequivalence and the inequivalence using one PK

 

      endpoint.

 

                We use the ratio of geometric means Mu                         

                                                                                

    T/MuR

 

      to define bioequivalence and INequivalence.  Mu                          

                                                                               

  T

 

      represent the geometric mean of the test product,

 

      and the Mu                                                R is the

geometric mean of the reference

 

      product.

 

                The bioequivalence is true when the ratio

 

      is between 80 percent and 125 percent.  We call

 

      this bioequivalence interval.  Outside this

 

      bioequivalence interval is defined as

 

                                                               225

 

      bioINequivalence region.

 

                Now, to test the bioINequivalence, the

 

      null hypothesis is the bioequivalence interval.

 

      The alternative is the bioINequivalence regions.

 

      Similar to bioequivalence test, we will perform

 

      two, 1-sided test, as well for bioINequivalence

 

      assessment.

 

                The null hypothesis for 1-sided test is to

 

      have the ratio reached and equal to 80 percent, and

 

      the alternative is less than 80 percent.  There is

 

      another test  that now is less than or equal to 125

 

      percent, and the alternative is driven 125 percent.

 

                We can claim bioINequivalence if one of

 

      the two nulls is rejected.  We perform the test on

 

      the significance level of 0.05.  Now, this is the

 

      equivalent to form 2-sided 90 percent confidence

 

      intervals.

 

                The criteria to claim bioINequivalence is

 

      when the 2-sided 95 percent confidence interval

 

      lies completely outside the bioequivalence

 

      interval.

 

                I would like to remind everybody here that

 

                                                               226

 

      using 2-sided 90 percent confidence interval for

 

      bioINequivalence test, the error rate is not always

 

      protected at 5 percent level.  This is different

 

      from bioequivalence test.

 

                If we have a reasonable conducted study,

 

      say, it's a 2-sequence and a 2-way crossover

 

      design, and the subject is more than 20, and the

 

      within-subject standard deviation is less than 0.7,

 

      then, we can safely control the error rate to 5

 

      percent, however, if the subject sample size is

 

      less than 20, we have large variance for the test

 

      statistics, then, the error rate may not always be

 

      controlled at 5 percent.  In this case, we might

 

      have to consider to use 2-sided 95 percent

 

      interval.

 

                Let's move on to the definition of the

 

      bioequivalence and the inequivalence using three PK

 

      endpoints.  As mentioned before, the three PK

 

      endpoints is Cmax, AUCt, and AUCinfinity.

 

                The definition of bioequivalence is the

 

      cubic region in this 3-dimensional diagram.

 

      Outside this cubic region will be the

 

                                                               227

 

      bioINequivalence region.

 

                For the criteria for bioequivalence

 

      assessment using three PK endpoints is to require

 

      all 3, 2-sided 90 percent confidence interval for

 

      the ratios of our geometric means has to be

 

      reaching the bioequivalence limit.

 

                Now, the error rate of wrongfully

 

      rejecting bioequivalence using this criteria is

 

      protected at 5 percent level.

 

                Now, for bioINequivalence assessment using

 

      three endpoints, we are looking for a strategy that

 

      can control the error rate of wrongfully rejecting

 

      bioequivalence at a rate of 5 percent.

 

                Also, we want to control the error rate

 

      under all correlation structures because we do not

 

      know the correlation structure of the three PK

 

      endpoints.

 

                Now, to develop those strategies, we

 

      assume the variances of test statistics are not

 

      large.

 

                Now, there is a common misconception when

 

      assessing bioINequivalence.  The common

 

                                                               228

 

      misconception is that to claim bioINequivalence,

 

      when one of the three PK endpoints satisfies the

 

      bioequivalence criteria, which is the 2-sided 90

 

      percent confidence interval is outside of the

 

      bioequivalence interval.

 

                Now, we will not accept this kind of

 

      approach to assess bioINequivalence because it will

 

      inflate error rate of wrongfully rejecting

 

      bioequivalence.  The error rate can be up to 15

 

      percent if three endpoints are independent, can be

 

      about 8 percent if the three endpoints are highly

 

      correlated.  We consider that approach is quite

 

      liberal.

 

                Now, people may want to think about we can

 

      use quite tough criteria, which is to claim

 

      bioINequivalence if all the three PK endpoints

 

      satisfy the bioINequivalence criterion, which is

 

      2-sided 90 percent confidence interval outside of

 

      the bioequivalence interval.

 

                These tough criteria will tightly control

 

      the error rate under all correlation structures,

 

      however, it won't provide good power to demonstrate

 

                                                               229

 

      bioINequivalence, therefore, we are not

 

      recommending to use this criteria either.

 

                What we would like to recommend are the

 

      following strategies.  The first strategy we

 

      present here is to pre-specify one of the three PK

 

      endpoints for bioINequivalence test.  For example,

 

      if you decided to use AUCt to test the

 

      bioINequivalence, then, you can perform analysis on

 

      this endpoint only, ignore the other two.

 

                The requirement for this strategy is that

 

      you have to pre-specify this endpoint in your study

 

      protocol.  Otherwise, you could end up switching

 

      endpoints, which may inflate error rate, which we

 

      don't like to see that.

 

                Now, this strategy is ideal for situations

 

      when one knows that one specific PK endpoint is

 

      more likely to show bioINequivalence than others,

 

      but it may have poor power if you misspecify the

 

      endpoint.

 

                Another strategy we would like to

 

      recommend is called Bonferroni corrections.  There

 

      are many versions of Bonferroni corrections.  One

 

                                                               230

 

      example of using Bonferroni correction is to use a

 

      2-sided 96.7 percent confidence intervals for three

 

      endpoints instead of 90 percent confidence

 

      interval.

 

                If one of the three 96.7 percent

 

      confidence interval fall in the bioINequivalence

 

      regions, we will say the bioINequivalence is

 

      demonstrated.

 

                This strategy is ideal for scenarios when

 

      one knows that one PK endpoint is more likely to

 

      demonstrate bioINequivalence than others, but do

 

      not believe that all the endpoints have good power

 

      to demonstrate bioINequivalence.

 

                Another strategy we would like to discuss

 

      here is to use three confidence intervals with

 

      different lengths.  This can be considered a

 

      variation to the approach that requires all the

 

      three endpoints to satisfy the bioINequivalence,

 

      which is the very tough criteria.  This criteria

 

      will control the error rate no more than 5 percent.

 

                One example of this criteria is to use 94

 

      percent confidence interval, 98 and 96 percent

 

                                                               231

 

      confidence interval for the three endpoints instead

 

      of all three, 90 percent confidence intervals.

 

                This approach is ideal for situations when

 

      one has no idea which PK endpoint has the best

 

      power, but you know that all the three endpoints

 

      could show bioINequivalence.

 

                To support what we have discussed for the

 

      three strategies, I would like to show you some

 

      power examples for several scenarios for the three

 

      strategies.

 

                We calculated power under two correlation

 

      structures.  The first scenario is that only one

 

      endpoint has good power to demonstrate

 

      bioINequivalence.  One example is that AUCt has

 

      only 5 percent power, AUCinfinity has 20 percent

 

      power, and Cmax has the best power, which is 90

 

      percent.

 

                In this case, if we choose pre-specified

 

      strategy, we could have a power between 5 percent

 

      and 90 percent, so this example clearly shows that

 

      if you know Cmax is the endpoint that could give

 

      you the best power to demonstrate bioINequivalence,

 

                                                               232

 

      then, you should choose the strategy to pre-specify

 

      Cmax in your protocol.

 

                In this example, Bonferroni correction

 

      also can give you quite a robust power, but the

 

      varying confidence interval approach is not as good

 

      as the other two approach.

 

                Now, for second scenario, which is all

 

      three endpoints have reasonable power to show

 

      bioINequivalence. Now, here, one example is AUCt

 

      has 60 percent power, AUCinfinity has 70 percent

 

      power, and Cmax has 80 percent power.

 

                Now, the per-specified approach give you

 

      60 to 80 percent power.  The Bonferroni correction

 

      will give you about 64 to 72 percent power, and

 

      under the varying confidence interval approach, we

 

      will give you about 70 percent power.

 

                So, in this case, if you feel that all

 

      three endpoints could demonstrate bioINequivalence,

 

      then, varying confidence interval approach might be

 

      a good choice.

 

                This scenario is that all three endpoints

 

      have equal power.  All has 80 percent power.  In

 

                                                               233

 

      this case, if you know exactly that all has 80

 

      percent power, then, you can choose pre-specified

 

      approach, but this is probably unlikely known to us

 

      before we do the experiment.

 

                Now, in this approach, Bonferroni

 

      correction will give you decent power, but varying

 

      confidence interval will give you probably the best

 

      power if you don't know that all the endpoints has

 

      equal power.

 

                This leads to the summary of our

 

      recommendations on using three PK endpoints for

 

      assessing bioINequivalence.  When one knows which

 

      endpoint is more likely to show bioINequivalence,

 

      Strategy I should be used, which is to pre-specify

 

      the endpoint in study protocols and use the

 

      endpoint to test bioINequivalence.  For this

 

      approach, you should use a two-sided 90 percent

 

      confidence interval.

 

                When one knows that one endpoint may have

 

      good power, but do not believe that all of them,

 

      all of the endpoints have good power, then, we

 

      suggest to use Strategy II, which is the Bonferroni

 

                                                               234

 

      correction.

 

                One example of Bonferroni correction is to

 

      use a two-sided, 96.7 percent confidence interval.

 

      If you believe that all three endpoints could have

 

      reasonable power to show bioINequivalence, then,

 

      Strategy III should be recommended.

 

                The example of Strategy III is to use 94

 

      percent and 98 and 65 percent confidence intervals.

 

                Thank you.

 

                DR. KIBBE:  Questions?  Go ahead.

 

                Committee Discussion and Recommendations

 

                DR. GLOFF:  Art suggested over lunch that

 

      I say something this afternoon to earn my keep

 

      here.  I have a question, it is probably an

 

      uninformed question, so I apologize for that.

 

                Is there any provision in all this to look

 

      at the results for the reference product relative

 

      to the results for the reference product that were

 

      obtained either by the company that submitted the

 

      ANDA in the first place or prior results submitted

 

      by the holder of the NDA?

 

                DR. LI:  To my knowledge, I don't think

 

                                                               235

 

      so.  I don't know.

 

                DR. YU:  I guess the bioequivalence

 

      confidence is defined 88 to 125 percent, so that is

 

      the criteria we use. Under certain circumstances,

 

      when, for example, the variability is significantly

 

      high, we will get clinical studies.  We will look

 

      at the availability, how much they impact the

 

      confidence interval, but the criteria still remains

 

      80 to 125 percent.

 

                DR. GLOFF:  Well, I am sure you probably

 

      have thought of why I am asking that question.  A

 

      prior speaker made the comment that when a generic

 

      is being developed and submitted, that they could

 

      hand-pick the lot that the generic company uses to

 

      compare to and hand-pick the lot of the innovator

 

      product.

 

                I am sitting here thinking, well, why

 

      couldn't the innovator company do the exact same

 

      thing to try to demonstrate that the two were

 

      bioINequivalent or were not bioequivalent, and I

 

      don't know if the Agency came to that conclusion,

 

      what they would then do for sure, but I am

 

                                                               236

 

      concerned about that, that you do that, and then

 

      where are you.

 

                DR. YU:  I guess the information for

 

      bioavailability, bioequivalence, as clinical

 

      pharmacology sections, it is available in the

 

      public domain, so any sponsors, any companies out

 

      there, you can request, go through Freedom of

 

      Information and get those information from FDA

 

      before you conduct any studies.

 

                DR. KIBBE:  Go ahead, Ajaz.

 

                DR. HUSSAIN:  I think the question that is

 

      being asked is how do we relate one study finding

 

      to what happened in the previous one.  I mean that

 

      is the fundamental question.

 

                We actually don't do that.  We often don't

 

      do that.  But I think that is an important point,

 

      and I have tried to look across different ANDA

 

      submissions, especially when Gordon Amidon was at

 

      FDA and we did a lot of the data mining for our BCS

 

      classification, we looked at all that.

 

                I think there is value to that, but often

 

      we find that absolute numbers that you see in terms

 

                                                               237

 

      of percentages is fine, but the absolute values

 

      that you see depends on the assay variability, and

 

      so forth, differences, and so forth.

 

                But I think no matter what you see, one

 

      study, the second study being done to show

 

      bioINequivalence in the first study, there is an

 

      aspect of selecting the thing, and that has been

 

      discussed as Marvin said, profoundly by the father

 

      of biopharmaceutics, Garrett Levy, so that is part

 

      of the systems.

 

                DR. KIBBE:  Jurgen.

 

                DR. VENITZ:  I have a question on Slide

 

      11.  This is where you were discussing using the

 

      three PK parameters and you are commenting that you

 

      don't recommend that because there is not adequate

 

      power.  I would like for you to explain that

 

      statement to me.

 

                DR. LI:  This criterion to require all

 

      three endpoints to show bioINequivalence, the

 

      bioINequivalence criteria for one single endpoint

 

      is 2-sided 90 percent confidence outside of the

 

      bioequivalence region.

 

                                                               238

 

                So, if you remember our previous talk, we

 

      showed a power of showing bioINequivalence.  It is

 

      pretty hard to show bioINequivalence for one

 

      endpoint.

 

                DR. VENITZ:  How do you define power?

 

                DR. LI:  Power is the probability to show

 

      bioINequivalence if bioINequivalence is true.

 

                DR. VENITZ:  So, you are ignoring the fact

 

      that you have a previous study that says the two

 

      products are bioequivalent, in other words, your

 

      power is only defined post hoc after this single

 

      experiment that you are trying to address?

 

                DR. LI:  No, the power is not defined by

 

      the experiment.  It is a probability which you

 

      don't know actually, you do not know.

 

                DR. VENITZ:  But you do have prior

 

      information that two products are bioequivalent,

 

      right?

 

                DR. LI:  Right, you could have.  What has

 

      driven power is how far the bioINequivalence away

 

      from the bioequivalence interval.  The further away

 

      from bioequivalence interval, you have back-up

 

                                                               239

 

      power, and also it depends on how many sample size

 

      you use, which is sample size basically reduce the

 

      variability.

 

                DR. VENITZ:  But don't you then ignore, as

 

      I said before, in your power, the way you define

 

      power, the fact that you have prior information?

 

      You are just basing it on a single experiment.  You

 

      already have an accepted study that says those two

 

      products are bioequivalent, and now you are saying,

 

      well, I need more power to show that they are

 

      bioINequivalent.  Isn't that kind of a

 

      contradiction?

 

                DR. LI:  I didn't see the contradiction.

 

      What I am trying to explain to you, that to use

 

      three endpoint to show bioINequivalence--

 

                DR. VENITZ:  Is hard.

 

                DR. LI:  --is harder.

 

                DR. VENITZ:  Right.

 

                DR. LI:  Because the power is lower.

 

                DR. VENITZ:  And I am saying you already

 

      have evidence to suggest that they are

 

      bioequivalent, shouldn't it be harder.

 

                                                               240

 

                DR. LI:  Well, no, if the drug is truly

 

      bioINequivalence, if you design your study

 

      properly, you should have good power to show that,

 

      but if you choose this criteria, you probably won't

 

      have good power.  You could choose the better

 

      criteria that give you better power.

 

                DR. KIBBE:  You are beaten.

 

                DR. VENITZ:  Okay.

 

                DR. KIBBE:  She is talking about

 

      statistical power of the individual study presented

 

      to her.

 

                DR. VENITZ:  I am talking about the

 

      overall power to rule whether something is

 

      bioequivalent or not in the totality of the

 

      information that you have, not just the specific

 

      study, which is what you are talking about.  You

 

      are talking about a specific study where you look

 

      at the three parameters individually, you correct

 

      it or all three of them.

 

                DR. YU:  I guess, Jurgen, you are

 

      absolutely correct.  Actually, we have many, many

 

      debates and discussions, Qian knows that, talk

 

                                                               241

 

      about when the statistic versus you have a prior

 

      knowledge about bioequivalence or bioINequivalence

 

      or quality.

 

                Certainly, what we are trying to address

 

      here is actually, you have five potential options.

 

      One of the options is you have no prior knowledge

 

      whatsoever.  That is one of them we have to present

 

      as a complete picture, we are not recommending

 

      this.

 

                One of the options, we say--all the option

 

      scenario out there, I guess, one of the scenarios,

 

      in your mind, you have a prior knowledge that is

 

      impossible, but for the completion of the picture,

 

      that was presented.

 

                DR. KIBBE:  We have got Ken and then Paul,

 

      Nozer, and me, and Marvin.

 

                DR. MORRIS:  A quick comment.

 

      Irrespective of whether this is innovator or

 

      generic, I think the fact that you can hand-pick

 

      lots that are this different says more about the

 

      process that is being used to make our products

 

      than it does our testing for bioINequivalence.  I

 

                                                               242

 

      think this was Levy's point either directly or

 

      indirectly is that it is probably more to the point

 

      that we need to control our processes to the point

 

      where Jurgen's observation becomes the rule in a

 

      sense.

 

                DR. KIBBE:  Paul.

 

                DR. FACKLER:  Part of the problem, I think

 

      is we are trying to do an exact science here where

 

      the whole issue is so variable, it is out of our

 

      control.  If we run the same study in the same set

 

      of subjects twice, we will get two different

 

      results, the same drug product, the same people,

 

      and it's different.

 

                The variability is just unmanageable, so a

 

      generic company will take a lot of innovator

 

      product and a lot of generic product and run it in

 

      a certain number of subjects, and that number is

 

      calculated to give us 80 percent power. Four out of

 

      five times, those products will statistically

 

      appear to be bioequivalent, and one out of five

 

      times they won't, they are not bioequivalent.

 

                It's statistics, and we should not spend

 

                                                               243

 

      too much time trying to get an exact measurement

 

      here of what bioequivalence is, nor what

 

      bioINequivalence is.  I think the question is

 

      really when is a product not going to perform for

 

      the patient who is taking it, and we have

 

      arbitrarily said 80 to 125 works, and there is some

 

      anecdotal evidence over the last 25 years that the

 

      generic products on the market work.

 

                So, I would just caution the committee to

 

      be careful defining when you would want to pull one

 

      of those products off the market.

 

                DR. KIBBE:  Nozer.

 

                DR. SINGPURWALLA:  Well, first is I think

 

      you have done a very thorough, detailed analysis

 

      given a badly defined problem.  You have done very

 

      well.  Thank you.

 

                Now, I am going to comment.  I am going to

 

      ask you two questions.  You have this AUCt.  How do

 

      you pick the t?

 

                DR. LI:  The t is the time point that you

 

      can still identify the drug concentration in your

 

      blood.

 

                                                               244

 

                DR. SINGPURWALLA:  It's the last.

 

                Is it possible that for one t, you will

 

      arrive at one decision, and for another t, you will

 

      arrive at another decision, which goes back to what

 

      Paul has been cautioning us about?

 

                DR. LI:  Uh-huh.

 

                DR. SINGPURWALLA:  That is one comment.

 

      The last comment is your last viewgraph.  When you

 

      say "recommendations," you have three bullets.

 

      When one knows this, you do this.  When one know

 

      this, you do this.  When one knows this, you do

 

      this.

 

                Well, what do you do when one knows

 

      nothing?

 

                DR. LI:  Actually, this is a very good

 

      point.  If you see my example, the 3 example, if

 

      you know nothing, actually, Bonferroni correction

 

      give you quite reliable robust power even it is not

 

      the best power for that situation, but it give you

 

      pretty good power, we might recommend this, and

 

      actually, this probably will be the default

 

      approach for us to review if the sponsor didn't

 

                                                               245

 

      specify any approach.

 

                DR. SINGPURWALLA:  So, maybe you should

 

      put a fourth bullet, if you know nothing, do this.

 

                But now let me go back to the main

 

      discussion that has been spawned by Carol's

 

      question.  I think both Jurgen and Ajaz have been

 

      dancing around the issue, and not coming out right

 

      and saying what is on their mind.

 

                Basically, what is on their mind is if you

 

      have prior information, which you do have, what do

 

      you do, and really what we should do is not address

 

      the problem in the manner in which this is

 

      addressed.  No criticism intended to you of the way

 

      you have done it.  You have done it very well.

 

                I think you should formulate this as a

 

      problem in making decisions.  You either declare

 

      bioequivalence or you declare non-bioequivalence,

 

      and the declaration of one or the other is a

 

      function of what risks it may entail if you make

 

      the wrong decision, so that takes care of Paul's

 

      argument that there is so much variability.

 

                You make decisions in the face of

 

                                                               246

 

      variability, so that would recast this whole

 

      problem, reformulate it, and readdress it.  It's a

 

      serious issue, because really, what you are all

 

      doing is building a superstructure on something

 

      that is not carefully defined.

 

                Thank you.

 

                DR. KIBBE:  Marvin, do you want to jump in

 

      or do you want me to jump in?

 

                DR. MEYER:  Well, let me just comment.  In

 

      my experience--and there is probably exceptions

 

      certainly--if I had to pick a parameter to show

 

      bioINequivalence, I would go with Cmax.  That tends

 

      to be a lot more variable, wider confidence limits,

 

      so I think you could probably go a priori with Cmax

 

      and use Strategy I if you wanted to do that.

 

                You are saying if I do that, then, the

 

      confidence limits has to be totally outside of 80

 

      or 125.

 

                DR. LI:  Right, yes.

 

                DR. MEYER:  Otherwise, you can't tell

 

      perhaps.

 

                DR. LI:  Yes, it is exactly the picture

 

                                                               247

 

      Lawrence showed you before, and if you feel

 

      uncomfortable, I think there is a second picture

 

      that has failed to show bioINequivalence.

 

                Actually, I would like to come to answer

 

      that question from a statistical point of view.

 

      You like to see, you know, the picture, if the

 

      confidence interval overlap to about bioequivalence

 

      interval, which is the second case.

 

                DR. MEYER:  I am really more worried about

 

      means.

 

                DR. LI:  The mean is outside the

 

      bioequivalence.  Let me tell you about the

 

      statistical concerns.  If we claim, be clear, this

 

      is bioINequivalence, then, you end up to make the

 

      error that is a modern FAQ [ph] event, which for

 

      statisticians, we do not like to see this happen,

 

      and if you have the confidence interval completely

 

      outside of the bioequivalence interval, then, we

 

      are comfortable that if you would claim this drop

 

      is bioINequivalence, we won't make error more than

 

      5 percent.

 

                I know for pregnancy test, you could claim

 

                                                               248

 

      that lady not pregnant, even 51 percent sure, and

 

      you could make a 49 percent error, but for

 

      statistical decision, for regulatory decision, we

 

      cannot make more than 5 percent error.  That is why

 

      we define it has to be outside the region.

 

                DR. MEYER:  I guess I worry about too much

 

      rigor in that.  Let's say the point estimate was 60

 

      percent, pretty bad, and the confidence limit,

 

      because of high variability, went over onto 80.2

 

      percent.

 

                You have a bioINequivalent product, there

 

      is no question about it.  You are not going to fix

 

      that by anything other than reformulation, but

 

      because of variability, you managed to slop that

 

      righthand tail over above 80.

 

                DR. LI:  What if people tell you the study

 

      is conducted using only five subjects, and you see

 

      the point estimate is 60 percent, and you have a

 

      confidence limit, you know, almost everywhere, can

 

      you claim this is bioINequivalence?

 

                DR. MEYER:  No, that is why I think you

 

      ought to fix N, too, to avoid that kind of an

 

                                                               249

 

      issue, and maybe even fix the mean must be less

 

      than the mean ratio in the ANDA.

 

                DR. LI:  If we fix, that will lead to

 

      stagnation [?] of bioequivalence and

 

      bioINequivalence.  Maybe we will fix our approach

 

      after the problem is redefined.

 

                DR. YU:  I do not see actually any

 

      difference.  I personally perfectly understand your

 

      concern.  For example, there are two scenarios we

 

      can talk about to this figure, which figure No. 2.

 

      One of the scenarios is point estimate is 79, the

 

      confidence interval is 78 versus 81.  Another

 

      scenario is the point estimate is 60, confidence

 

      interval is 40 to 80, for example.

 

                Under these two scenarios, from

 

      statistical perspective, we cannot give you a

 

      definitive answer, however, the first scenario, do

 

      you know the drug.  Certainly, we can definitely

 

      use prior knowledge with respect to safety and

 

      efficacy of this drug, and we will make a

 

      scientific decision.

 

                The first case, you might have to think

 

                                                               250

 

      about it, because this drug is still on the market,

 

      you are perfectly okay, and you will make a

 

      scientific decision on the second case.  Obviously,

 

      we will not close eyes and say let it go,

 

      definitely not, and chance to be pulled to the top

 

      of the company is high.

 

                Even the first case, we will inform the

 

      company, we will discuss with the company what to

 

      do with that case.  Certain case, the probability

 

      to be pulled is higher.  I would not say 100

 

      percent definitely as the first case, that

 

      bioINequivalence case, this case, certainly we will

 

      take a look at it and discuss it with our

 

      clinicians within FDA, discuss it with our sponsors

 

      outside of FDA to take a proper action as issues

 

      occur.

 

                I hope this answers your question.  Thank

 

      you.

 

                DR. KIBBE:  Let me just throw a few random

 

      thoughts in on the table with the intention of

 

      keeping us all past our flights, so everybody

 

      misses their flight.

 

                                                               251

 

                First, if a product has already been

 

      established as bioequivalent, it has been on the

 

      market for a while, and we have a lot of confidence

 

      in the product, then, I think to pull the product

 

      off the market, we have to make a clear and

 

      distinct argument that the product is indeed

 

      failing to live up to the criteria that was

 

      established for it.

 

                It is hard for me to imagine a product

 

      that got on the market with a bioequivalency study

 

      where the mean values were, say, 97 percent or 103

 

      percent of the mean, the innovator, and well within

 

      the confidence interval, and all of a sudden you

 

      are going to find a lot that is going to be a

 

      disaster.

 

                However, it is possible.  If that is the

 

      case, then, you have before you two experiments

 

      with opposite results, and in most laboratories

 

      that I have been involved with, when you have two

 

      experiments with opposite results, everybody looks

 

      at each other and says we have got to do it again,

 

      we can't just leave it like this.

 

                                                               252

 

                So, no matter what you do as an agency

 

      setting up guideposts for the innovator to come

 

      forward with a bioINequivalency study, I think then

 

      the Agency says thank you very much for a second

 

      piece of information, and we now must resolve the

 

      discrepancy, not by trumping their study with your

 

      study, or trumping your study with their study, but

 

      doing the critical study, which is now the Agency

 

      should go out in the marketplace and buy 100 of

 

      each of the products off the shelf somewhere, maybe

 

      St. Louis, maybe Kansas City, somewhere.

 

                I would be afraid that if we went to

 

      Canada, we would get much higher quality products,

 

      and we want to stay with the quality level here,

 

      and do the third study, and then say, okay, your

 

      company did it with whatever biases that might have

 

      been involved in the selection to get on the

 

      market, and your company did it with whatever

 

      biases or not that you had and to show that it was

 

      off market, and we did it, and now we have the

 

      definitive result, and we have to limit the number

 

      of times you can come forward and do this with us.

 

                                                               253

 

                So, we have done the third study.

 

      Otherwise, I think it is really one of those he

 

      trumps you, and you trump him, and if they come

 

      forward with a bioINequivalency study that seems to

 

      pass the criteria, whatever you pick, and I come

 

      and give you a second bio study with the product

 

      and say, look at that, it really is good, what do

 

      you do?

 

                Let them trump and trump and trump, and I

 

      know the CROs are all saying oh, shut up, let them

 

      do it, because we can do these studies, you know,

 

      once a month, it would be okay, but it is not going

 

      to get you the final answer.

 

                There is a couple of other things that you

 

      might want to keep in the back of your mind.  Drugs

 

      which are non-linear, are easy to manipulate.

 

                If you do a study with dilantin at 50

 

      milligrams per patient, and you get a bioequivalent

 

      result where one is just slightly higher than the

 

      other, then, get a group of patients and give them

 

      400 milligrams, and you will run that mean right

 

      off the table, and what would be an insignificant

 

                                                               254

 

      difference at a reasonable therapeutic level would

 

      not be a insignificant difference at an elevated or

 

      above normal therapeutic level.

 

                There is lots of things we can do, so that

 

      if we are going to get into this, I would go with a

 

      nice tight bioINequivalent study, and we can argue

 

      the value of the statistics, but that can't be the

 

      end.  That absolutely cannot be the stopping point.

 

                That is just one more piece of data, as

 

      Jurgen correctly points out.  We already have data,

 

      now we have new data, and to resolve it, we have to

 

      have an impartial arbiter, and the Agency has to do

 

      its own biostuff.

 

                Gordon wants to disagree with me.  Go

 

      ahead.

 

                DR. MEYER:  Actually, I think that is a

 

      good idea, but I think that given the resources,

 

      that there is no reason that an innovator can't be

 

      expected to do a study properly.  They will get

 

      inspected on the first one anyway,  the first

 

      bioINequivalence study, they can be inspected on

 

      the second one.  They are not going to risk their

 

                                                               255

 

      reputation by messing around with the data, so I

 

      don't think the Agency, I mean that would be

 

      impractical for the Agency to have to run out and

 

      do a confirmatory study.

 

                DR. YU:  We come back the April 14th

 

      discussion, and I think all of us heard Gary made

 

      the presentation back in July that our submissions

 

      this year increased 25 percent, and then we talk

 

      about risk management, we talk about where we put

 

      our resource in, and all we are doing for this

 

      bioINequivalence type is Agency have defined the

 

      criteria for bioequivalence, we want to define the

 

      criteria for bioINequivalence to make a

 

      clarification out there.

 

                That's all come back because in the

 

      literature, scientific literature, people tend to

 

      conduct a bioequivalence study, now the confidence

 

      interval is 79 or 126, claim as bioINequivalence.

 

                We say this is not scientifically valid.

 

      So that is the whole purpose is wanting to give a

 

      clear definition with respect to bioequivalence

 

      versus the bioINequivalence, as well as a fail to

 

                                                               256

 

      demonstrate bioequivalence and a fail to

 

      demonstrate bioINequivalence.

 

                Then, from here, you are absolutely

 

      correct, when we see a study like that, if we

 

      cannot--we are trying to put our resources in the

 

      NDA reviews.  Just in case this happened, cannot be

 

      very clear, there is an ambiguity in the gray area,

 

      certainly, Agency will have to put the resource

 

      whether we like it or not.  I think we agree.

 

      Thank you.

 

                DR. KIBBE:  Go ahead, Gordon.

 

                DR. AMIDON:  I am not going to disagree

 

      with you, Art, but the question I have regarding

 

      the scenario where a product is bioequivalent and

 

      on the market, and another company comes in showing

 

      potential bioINequivalencies, has the product

 

      changed.

 

                If we had a good dissolution criteria in

 

      evaluation, we would have some underlying possible

 

      more scientific hypothesis to make rather than run

 

      out and test another set of products.

 

                So, I think it comes down to dissolution.

 

                                                               257

 

                DR. KIBBE:  Just so you know, bayesian

 

      dissolution.

 

                DR. GLOFF:  One quick comment on what

 

      Gordon just said.  He said has the product changed,

 

      and my question would be, and if so, which of the

 

      two products has changed.  It is not necessarily

 

      just the generic.

 

                MR. BUEHLER:  Well, Lawrence made a good

 

      point, and this is a resource issue for us.  We

 

      haven't gotten that many challenge studies

 

      recently.  When we do get them, they are usually

 

      out by, like Lawrence said, a little, 127, 79,

 

      something like that, but the letters that accompany

 

      them are very profound.

 

                They are big public health issues, they

 

      are always presented as huge issues, and, of

 

      course, we have to look at these, we have to

 

      address the issue and resolve the issue because I

 

      agree with Gordon, we don't want generic products

 

      out there that are bioINequivalent.  That is a

 

      problem for that particular product, it's a problem

 

      for the entire industry to have products where the

 

                                                               258

 

      American public can't have confidence in those

 

      particular products.

 

                We want to know about those products, and

 

      we want to know when products are truly

 

      bioINequivalent, but we don't want to have to deal

 

      with all of these studies that come that are just a

 

      hair out one way or a hair out the other.

 

                We like rules in the Office of Generic

 

      Drugs, and we are sort of bound by our rules, and

 

      however, you know, they are criticized by some

 

      statisticians, we do have our 80 to 125 rule, and

 

      we stick by that very rigidly, and to not do that

 

      would mean a tremendous creep, you know, a

 

      tremendous I think lack of confidence in the

 

      generic process.

 

                Is 79 okay?  Well, you know, sure, okay,

 

      79 is okay.  But what about 78, what about 77?  You

 

      can to down, you can go up, and the next thing you

 

      know, you have a confidence interval you can drive

 

      trucks through.

 

                So, that is why we are very particular

 

      about rules, and in this particular case, you know,

 

                                                               259

 

      this is what we are trying to get for

 

      bioINequivalence, is some kind of a rule that

 

      companies won't send these in if they know that we

 

      are not going to deal with them.

 

                If they know that our rule is it has got

 

      to be this far out or this far over--

 

                DR. KIBBE:  Let me ask you a question.

 

      The ones that you have gotten, would they have

 

      passed this rule that you are putting--

 

                DR. YU:  The one we have right now--how

 

      many addition we have?  Probably 41 additions

 

      already back and forth, four or five people

 

      involved with the lawyer and the scientist

 

      involved.  The case we have, we hope we resolve

 

      very soon, but this case submitted to us back to

 

      '99, I think, submitted again in 2002, and you can

 

      see how many resources we are putting in, more than

 

      two years already passed.

 

                The issue is this case we are here, the

 

      confidence interval is--I have got a lower one--the

 

      top one is 126.

 

                DR. KIBBE:  And it's Cmax.

 

                                                               260

 

                DR. YU:  It's Cmax.

 

                DR. KIBBE:  So Marvin is right.

 

                DR. YU:  Oh, it's always Cmax.

 

                DR. KIBBE:  Of course, it is.  Would this

 

      proposed rule have said upfront that you haven't

 

      established your case?

 

                DR. YU:  Absolutely, yes.  You can see

 

      that, two people for two years.

 

                DR. VENITZ:  Can I make an observation and

 

      then give my recommendation?  First, I agree with

 

      Nozer, we are trying to squeeze a bayesian problem

 

      into a frequentist scenario, but given the fact

 

      that we have been hearing this time and last time

 

      that those are the rules that have been in effect

 

      for 20-plus years, that we don't find people dying

 

      on the streets, or they might be working actually

 

      in terms of providing safe and effective generic

 

      products, you are stuck with the system the way it

 

      is right now.

 

                So, I way I tried to approach it, not

 

      being a statistician, we have a body of evidence to

 

      suggest that the generic and the reference product

 

                                                               261

 

      are bioequivalent.  That is the reason why it got

 

      approved in the first place.

 

                I assume as part of your review, you are

 

      going to look for things that might have changed,

 

      creep in either the product or the reference

 

      product.

 

                So, now you have a claim being made that

 

      seems to contradict that, and then, in my mind, the

 

      burden of proof is with the person or the

 

      organization that files that claim.  So, the burden

 

      of proof to me means that it has to be difficult

 

      for them to overcome what you already know, so I

 

      personally would go with the toughest off your

 

      recommendation, and you have excluded my favorite

 

      one, which is all three of them have to pass:

 

      Cmax, AUCt, and AUCinfinity have to pass, because

 

      that is toughest route.

 

                If you can overcome that, then, I think

 

      you can argue, well, that is about as much evidence

 

      as you need given the fact that you already had

 

      pre-existing bioequivalence.  Then, you have enough

 

      to overrule.

 

                                                               262

 

                So, I would recommend what you didn't

 

      recommend, that all three parameters, all three

 

      metrics have to pass in order to conclude

 

      bioINequivalence.

 

                DR. KIBBE:  What do you recommend, Marvin?

 

                DR. MEYER:  I might say I am going to

 

      apologize for not being a statistician--I once said

 

      pharmaceutical scientists all apologize for not

 

      being statisticians, but statisticians don't

 

      apologize for anything.  I think that probably

 

      applies here.

 

                DR. KIBBE:  I will let you comment on that

 

      later, Nozer.

 

                DR. MEYER:  Under Strategy I, Lawrence,

 

      prespecify one of the three PK endpoints, and then

 

      analyze that.  Now, if you do a PK study, you are

 

      going to have all three at hand.  Why don't you

 

      just do Cmax and then do AUC, and then do

 

      AUCinfinity, and look at the data?  What is the

 

      problem with doing that?

 

                DR. YU:  Let me explain that first, that

 

      the criteria, when we define, statistically

 

                                                               263

 

      significant or not, is 5 percent of criteria.  If

 

      below 5 percent, statistically significant; above,

 

      it is difficult to say.

 

                For you to not prespecify anything, you

 

      conduct a study, the chance to be wrong is higher

 

      than 5 percent.  In fact, change on one slides, I

 

      believe it could be high, like 14 percent.

 

                DR. MEYER:  Isn't that if you use all

 

      three?

 

                DR. YU:  If you use any of three.  You

 

      don't not prespecify any of them, they are just

 

      looking for one.  For examples, these are the

 

      slides, the error rate could be 14.7 percent.  If

 

      that is the case, scientific speaking, is too high.

 

      Certainly, scientific speaking will look at a case,

 

      you just present it and make a scientific decision.

 

                DR. MEYER:  But if you pick one and pick

 

      the wrong one, you also have a chance of being in

 

      error, which isn't in there somewhere.

 

                DR. YU:  That is correct.  You are

 

      absolutely correct, Marvin.  Actually, you

 

      understand very well in my judgment.  If you

 

                                                               264

 

      prespecify and if you use AUC, the wrong one, you

 

      could have a probability power.  The power could be

 

      5 percent to 90 percent, however, you just said you

 

      have a prior knowledge, so most likely you have

 

      probably picked the correct one.

 

                Now, let's put the stack back.  Indeed,

 

      there is a company out there.  Pick the wrong one,

 

      but even the wrong one you pick, for example, you

 

      pick the Cmax.  The case I will talk about is very

 

      theoretical.

 

                You pick up a Cmax, but it end up an AUC,

 

      you show the confidence interval, for example, 60

 

      to 79.  This is certainly the case is,

 

      statistically speaking, you do not see, the error

 

      could be a lot higher, but this does not mean we

 

      are going to close eyes, the Agency will not take

 

      an action, probably not, absolutely not.

 

                We certainly will investigate.  As we

 

      said, we look at a formulation change for both

 

      innovator and the generic side.  We look at all the

 

      scenarios.  We will have a bunch of people sitting

 

      in the conference for many hours, probably many

 

                                                               265

 

      meetings, and discuss with many parties and trying

 

      to make the best decision for the public.

 

                DR. MEYER:  Let me just say that if you

 

      pick an area under the curve, you are not getting

 

      any information about rate, because area under the

 

      curve can be quite independent of rate.

 

                If you pick Cmax, you are getting

 

      information about rate and amount, and also

 

      information about how different your population is,

 

      and lots of that kind of information, because Cmax

 

      has bounced all over the place especially when Cmax

 

      has also moved around Tmax, and the Tmaxes aren't

 

      constant, so your Cmax from one patient is going to

 

      be happening at half an hour, and then the next

 

      patient's Cmax is going to be happening at one

 

      hour, and you are going to have lots of fun.

 

                I really think Jurgen is right, that you

 

      need to establish a criteria that looks at all of

 

      the three parameters for bioINequivalency, because

 

      we look at all three parameters for bioequivalency,

 

      and pre-existing information has to be trumped

 

      effectively.  I still like the idea of doing a

 

                                                               266

 

      third study.

 

                DR. YU:  Thank you.

 

                DR. KIBBE:  Nozer, do you want to comment

 

      on his statistics?

 

                DR. SINGPURWALLA:  Oh, he was absolutely

 

      brilliant.  I am disappointed he went into

 

      pharmacy.

 

                DR. KIBBE:  Pat, go ahead.

 

                DR. DeLUCA:  In your diagram, Lawrence,

 

      you know, to me, there is only one here that

 

      demonstrates bioequivalence, the others are not

 

      bioequivalent, so however you turned them, the

 

      other four are not bioequivalent.

 

                But the question I would ask, in

 

      determining that the product was bioequivalent, you

 

      need Cmax and area under the curve.

 

                DR. YU:  Correct.

 

                DR. DeLUCA:  If they came in with just

 

      Cmax or just area under the curve, you wouldn't

 

      have approved that as being bioequivalent, is that

 

      right?

 

                DR. YU:  Absolutely.

 

                                                               267

 

                DR. DeLUCA:  So, if they just had one that

 

      wouldn't be, you wouldn't approve it if they just

 

      had one. So, I can't see why, then, if you are

 

      looking at a product that is bioINequivalent, why

 

      you can't just use Cmax.

 

                You can just use one of them to me,

 

      because they had a pass, both of them, at the

 

      start, so if they didn't have both of them, they

 

      wouldn't have passed bioequivalence, so why isn't

 

      one enough?  Why isn't just Cmax enough to show

 

      bioINequivalence?

 

                DR. KIBBE:  Paul.

 

                DR. FACKLER:  Just one quick point.  The

 

      generic has to pass Cmax, AUC zero to t, and AUC

 

      zero to infinity under fasting conditions, under

 

      fed conditions, and for capsule beaded products,

 

      under sprinkle conditions.

 

                So, for some products, it is nine

 

      parameters that need to pass, for others, it is

 

      six, and for a relatively small group of products,

 

      it is three.  I just put that out there for what it

 

      is worth.

 

                                                               268

 

                DR. KIBBE:  Marvin.

 

                DR. MEYER:  You asked for a

 

      recommendation.  In the spirit of harmonization, I

 

      would suggest that Lawrence's figure there is

 

      perfect, that under standard conditions, no

 

      monkeying with the confidence limits, not 86, not

 

      three different ones, keep our 80 to 125, 90

 

      percent, and declare the two bottom ones

 

      bioINequivalent, and therefore bad, and therefore

 

      need investigation, and the rest of them are all

 

      either unknown or bioequivalent.

 

                DR. YU:  Thank you.  That is actually what

 

      we are recommending.

 

                DR. KIBBE:  Anybody else want to jump in

 

      on the consensus recommendation wagon?

 

                What do you think, Carol?

 

                DR. GLOFF:  I have a question for Marvin.

 

      Do you mean that all three parameters need to fall

 

      outside?

 

                DR. MEYER:  No, this could be any of the

 

      three parameters, any one of the three exhibiting

 

      either of the two bottom ones, bioINequivalence.

 

                                                               269

 

                DR. SINGPURWALLA:  I would like to make a

 

      comment from mathematics.  To disprove a theorem,

 

      all you need is one counter example, so if you want

 

      to show bioINequivalence, all you need is one

 

      violation.  If you want to show bioequivalence,

 

      then, you may have to go and do everything else.

 

                Does that rhyme well with your view,

 

      Jurgen?

 

                DR. VENITZ:  I am not sure because I still

 

      think that the hurdles that you have to overcome to

 

      get an approved generic on the market, not just

 

      looking at Cmax, I mean the other things as you

 

      have heard, and it may just not be a single Cmax,

 

      it may be other things.

 

                Given the fact that, as you have heard me

 

      talk about earlier this year, the 80 to 125 percent

 

      is really an arbitrary goalpost.  I do believe that

 

      the burden of proof should be high for somebody to

 

      get this reversed, to get an approval reversed.

 

                DR. SINGPURWALLA:  But the burden of this

 

      proof need not be so high.

 

                DR. VENITZ:  Think about what

 

                                                               270

 

      bioequivalence means.  It basically means you don't

 

      have enough evidence to reject a null hypothesis,

 

      to use statistical lingo.  You are basically trying

 

      to prove the impossible.  You can never prove that

 

      something is equal.  So, you are just bounding.

 

      You are saying, in my mind, I put arbitrary bounds

 

      on, and say, well, as long as it fits those bounds,

 

      we consider it to be bioequivalent.

 

                So, to disprove that, I think you have to

 

      disprove it on all the three metrics, the metrics

 

      that you used in the first place, to get approval.

 

                DR. KIBBE:  The argument that you are

 

      making is that because the criteria says that all

 

      three of these parameters have to meet the criteria

 

      to get approval, doesn't necessarily mean that we

 

      shouldn't require all three to meet the criteria to

 

      get unapproval.

 

                By saying okay, in the original

 

      submission, all they had to do is fail one to not

 

      get approval, that's fine, but now we have an

 

      already approved product, and we are doing a test

 

      to show that it is not equivalent, so I would like

 

                                                               271

 

      to see it demonstrated that it is not equivalent on

 

      the same three parameters, and if it can't do it on

 

      all three, then, it has failed that test, just like

 

      it would have failed originally to get approval by

 

      failing one of the three, and that is my argument.

 

                DR. SINGPURWALLA:  Off the record.  To

 

      really look into this issue, it is a much more

 

      serious issue that what meets the eye.  There is a

 

      rule that has been set up, and you have to live

 

      with that rule, I agree with you, but what is to

 

      stop the FDA from looking to the future and

 

      changing the rules?

 

                DR. HUSSAIN:  I think that point is well

 

      taken, and, Helen, actually that is exactly what my

 

      new instructions were from her, so we will take

 

      this further into discussion, and so forth.

 

                I think this was very valuable, and I am

 

      not fully sure exactly that we have come to a

 

      conclusion on this yet.

 

                DR. KIBBE:  Jurgen and I have come to a

 

      conclusion.  Marvin's conclusion is slightly

 

      variant, but not too much.

 

                                                               272

 

                DR. MEYER:  But I am retiring.

 

                DR. YU:  I guess that we are back to the

 

      April 14th situation where there is 1 versus 3.

 

                DR. HUSSAIN:  Lawrence, I think it is time

 

      to stop the discussion.

 

                DR. KIBBE:  I think the Agency has to step

 

      up to the plate.  We have given you the best advice

 

      we can.

 

                DR. YU:  Okay.  Thank you.

 

                DR. KIBBE:  I think it is appropriate at

 

      this stage, since my schedule says we are taking a

 

      break, to take a break.

 

                We are breaking 15 minutes early.  We will

 

      give you 10 minutes.  We expect you back in your

 

      seat at three minutes to 3:00, and at 3 o'clock, we

 

      will have our discussion about the locally acting

 

      gastrointestinal materials, and we will wrap that

 

      up in short order because Gordon has the exact

 

      answer we need right here.

 

                [Recess.]

 

                DR. KIBBE:  Ladies and gentlemen, the

 

      clock on the wall says it is three minutes to 3:00,

 

                                                               273

 

      and as it is my tradition, I will remind you to

 

      gather and begin.

 

                We have one more topic area.

 

                Lawrence, are you going to set the topic

 

      up?  You have got three minutes.

 

                DR. YU:  Yes, I will.  Actually, I can

 

      finish within two minutes.

 

               Bioequivalence Testing for Locally Acting

 

                         Gastrointestinal Drugs

 

                           Topic Introduction

 

                DR. YU:  The bioequivalence testing for

 

      locally acting drugs was I think presented to you,

 

      and I saw the comments.  Well, today, we are going

 

      to discuss the real issue of bioequivalence testing

 

      for locally acting drugs.  I will introduce this

 

      topic, and Gordon from the University of Michigan

 

      will give the talk on Scientific Principles, and

 

      Robert Lionberger from the Office of Generic Drugs

 

      will give you specific examples.

 

                Again, bioequivalence is defined as the

 

      absence in the rate and the extent of drug

 

      absorption.

 

                                                               274

 

                As I said yesterday, the pharmacokinetic

 

      measure for bioequivalence method for systematic

 

      drugs is well understood, well used.  We have

 

      pulled many, many products.

 

                The issue remains for locally acting

 

      gastro and GI drugs.  The part of reason for that,

 

      because the plasma concentrations may not be

 

      relevant to locally delivery bioequivalence, for

 

      example, a topical, nasal, inhalation, which I

 

      presented to you yesterday.

 

                The point we want to make sure that the

 

      dissolution controls the delivery to the site of

 

      action, whether it's the jejunum, jejunal ileum, or

 

      colon.  The drug concentrations in plasma are

 

      downstream from the site of action, unlike for

 

      systematic drugs, the drug ending systematic first,

 

      then, get the side effects action, for example,

 

      heart and liver, and so on, the heart and the

 

      brain.

 

                For GI acting drugs, there is no alternate

 

      absorption path because already they have to be

 

      absorbed from the gastrointestinal tract.  So,

 

                                                               275

 

      bioequivalence approach the Agency has used, for

 

      example, the clinical study of vancomycin,

 

      pharmacokinetics study is sulfasalazine, the in

 

      vitro study is cholestrylamine.

 

                What we want is want to develop a

 

      scientific basis for the choice of BE method,

 

      bioequivalence method, which we need your input on

 

      role of pharmacokinetic studies, role for in vitro

 

      dissolution studies, role of the clinical studies.

 

                With that short introduction, Mr.

 

      Chairman, I finished it within two minutes, I turn

 

      the podium to Gordie.

 

                DR. KIBBE:  Thank you, Lawrence.

 

                Dr. Amidon.

 

                         Scientific Principles

 

                DR. AMIDON:  Thank you.  I am glad we are

 

      recovered from that discussion of statistics.  I

 

      know I get glassy-eyed.  I think Qian Li did a

 

      great job, and then we turned it into chaos, but

 

      that is our job, I guess.

 

                MR. CLARK:  Chaos theory is our goal.

 

                DR. AMIDON:  What you received in the

 

                                                               276

 

      handout was the unedited version of my

 

      presentation, because it was done before I knew

 

      what Lawrence and what Rob were doing, so I am

 

      going to skip a lot of the slides that I have

 

      because points are already being made, and talk

 

      about the highlights, the essentials of my point,

 

      and I will give you the executive summary right

 

      now.

 

                First, bioequivalence is the question of

 

      dissolution.  What else is it?  The same drug in

 

      different products.  Once the drug is absorbed, it

 

      is the same except in the unlikely scenario, there

 

      is maybe a competitive metabolism inhibitor or an

 

      excipient that might alter permeability.  Evidence

 

      for that is limited in vivo in humans.

 

                So, bioequivalence is a question of

 

      dissolution.  That is where the science needs to be

 

      done.  So, the bottom line for GI drugs, for all

 

      drugs, is that we should put more emphasis on the

 

      science of dissolution and what I think of as a

 

      bioequivalence dissolution test.  So, that is going

 

      to be my bottom line, big picture conclusion.

 

                                                               277

 

                The subconclusion for GI drugs is that I

 

      think what we need is a bioequivalence dissolution

 

      test with some type of in vivo test, perhaps not a

 

      confidence interval test, maybe a point estimate

 

      and an interval requirement, say, between 90 and

 

      110, so that we don't have this confidence interval

 

      issue.

 

                You could argue do we need the in vivo

 

      test.  I think in some cases, we do not, and

 

      probably we need to go drug by drug for locally

 

      acting drugs, and Rob will talk about specific drug

 

      examples.

 

                One of the issues in setting up a policy

 

      issue is try to be very general, and you get into

 

      trouble because some things aren't generalizable

 

      very easily.  So, I think we will have to regulate

 

      GI drugs, of which there may be a half a dozen that

 

      are very important, more on a drug-by-drug basis,

 

      or maybe classify them, I don't know.

 

                So, that is the bottom line.  Dissolution

 

      is what we should be looking at, and a dissolution

 

      test plus an in vivo test, perhaps a point estimate

 

                                                               278

 

      would be enough.

 

                I am going to skip over most of these

 

      slides, because the points are already being made.

 

                The one point that I will make, that I

 

      make all the time, and I think is generally

 

      accepted, at least no one has argued, is that I

 

      think bioequivalence is maybe the single most

 

      important regulatory standard for virtually all

 

      products on the market today.

 

                That is, products on the market today, are

 

      on the market because of either proven or assumed

 

      bioequivalence.  If not, what could we say about

 

      the clinical?  We have to make that connection, the

 

      connection between the product in the bottle and

 

      the label is bioequivalence.

 

                Yes, of course, we have to have the

 

      potency, the impurities, and we have to have the

 

      standards, but bioequivalence, so this is I think

 

      one of the most important issues in drug regulatory

 

      standards in the world today, because it pertains

 

      to all products.  My interest, of course, is in

 

      oral products.

 

                                                               279

 

                There is some caveat in the Orange Book,

 

      just to point out kind of the legal basis.  If you

 

      look at where I think the most up-to-date

 

      definition of bioavailability and bioequivalence,

 

      it is in the preface to the Orange Book.

 

                It has been revised periodically over the

 

      years, I think no one has noticed it, because they

 

      slip kind of changes into the Orange Book, it just

 

      comes out, and life goes on.  Right?  Is that what

 

      you do?

 

                At any rate, for locally acting drugs, it

 

      says, "Where the above methods are not applicable,

 

      e.g., for drug products that are not intended to be

 

      absorbed into the bloodstream, other in vivo or in

 

      vitro test methods to demonstrate bioequivalence

 

      may be appropriate."

 

                That is where we are at here with GI

 

      locally acting drugs.

 

                Again, I am going to skip through these.

 

      You have seen this.  Rob is going to use this, so I

 

      am going to skip it, the disconnect for locally

 

      acting drugs.

 

                                                               280

 

                Now, classically, we do Cmax, AUC, of

 

      course, and AUCt, and we have confidence interval

 

      test, but if the levels in blood are very low, we

 

      have a problem, so we have a practical problem.

 

                So, I am going to come back to the

 

      paradigm for bioequivalence.  I think of the

 

      paradigm of bioequivalence today as being the

 

      following, starting at the top.  Similar plasma

 

      levels, similar pharmacodynamics, similar efficacy

 

      to the label.  I mean that is the implication,

 

      similar pharmacodynamics.

 

                Then, similar in vivo dissolution, similar

 

      plasma levels.  For oral products, I think maybe

 

      for all products, but certainly for oral products,

 

      similar in vivo dissolution.  When we think about

 

      the physiology of oral absorption, the drug

 

      dissolves and is spread along the gastrointestinal

 

      tract and absorbed.  We think of absorption as into

 

      the intestinal mucosal cell.

 

                Subsequently, systemic availability is

 

      later, and there is more stuff, drug stuff, you

 

      know, liver in between. That is part of the

 

                                                               281

 

      complication, we are doing bioequivalence based on

 

      plasma levels, which is systemic availability.  We

 

      are doing a systemic availability test which is

 

      distant for GI drugs from the site of action.

 

                So, similar in vivo dissolution, similar

 

      plasma levels.  So, then, where the science is

 

      today is in vitro dissolution.  We need to be more

 

      rigorous in how we do dissolution when we use it

 

      for bioequivalence materials.

 

                We think that there is no reason why we

 

      cannot establish better dissolution methodologies

 

      that reflect the in vivo dissolution process.  I

 

      think that would have a number of implications

 

      including accelerating the drug development

 

      process, because when you are making a product and

 

      then testing it in humans, you would like to have a

 

      good idea you are going to succeed.

 

                In order to do that, you have to have a

 

      dissolution test that reflects what is going on in

 

      vivo.  So, I think better dissolution can be a big

 

      step in advancing and accelerating drug

 

      development.

 

                                                               282

 

                So, that means doing something.  Often now

 

      today, we have what we call biorelevant dissolution

 

      media or biorelevant dissolution.  I think we need

 

      to use that term carefully because, you know, to

 

      take some natural surfactants and a little bit of

 

      phospholipid and put it in water and shake, you

 

      either have a drug delivery company or you call it

 

      biorelevant dissolution media, but what is it?

 

      There is no evidence that it is relevant to the in

 

      vivo dissolution process.

 

                So, we need to establish that connection

 

      between the in vitro dissolution methodology and

 

      the in vivo dissolution process, and I think that

 

      is where there is a big gap in our knowledge today,

 

      not just for GI drugs, for all drugs.

 

                So, my point here is broader than just GI

 

      drugs, but similarly, if we had confidence in an in

 

      vitro dissolution test, that is all we need to do.

 

      That is all we need to do.  So, we should be

 

      focusing the science on that dissolution test.

 

                I am preaching too much here, so I am

 

      going to skip most of my slides, but I have to show

 

                                                               283

 

      at least an equation.  I noticed that on my badge,

 

      they had MA.  It took me a while to remember that I

 

      had a Masters in mathematics, and I was impressed.

 

                The FDA is so thorough in their

 

      investigation of my background, you know, every

 

      year I have to fill out all these conflict of

 

      interest things.  They actually put MA.  That is

 

      the first time that has ever happened, so I have to

 

      compliment the FDA and their thoroughness in

 

      investigating my background.  But I did pass, and I

 

      am here.

 

                Anyway, this is the equation of

 

      bioequivalence, but I am not going to talk about

 

      it.

 

                We talk more about the physiology of

 

      gastrointestinal tract and product disintegration,

 

      dissolution, and spreading along the

 

      gastrointestinal tract. That is where the

 

      investigation is.  That is where we need to do more

 

      investigation.

 

                Again, I am skipping most of the slides.

 

                So, to kind of come to the conclusion on

 

                                                               284

 

      the bioequivalence for locally acting drugs.  I

 

      mean obviously, plasma level is downstream from the

 

      site of clinical effect, which is local.  The local

 

      site of action is in the GI tract.

 

                So, dissolution and transit in vivo

 

      controls the presentation of drug to the site of

 

      action.  So, this is where plasma levels are

 

      probably less good than a good dissolution test.

 

                Now, we could, with intubation, measure

 

      concentrations along at least part of the

 

      gastrointestinal tract, not easy to do, and, yes,

 

      you have got tubes in, so it is not normal.  You

 

      could argue is that feasible.

 

                Now, I think most locally GI drugs are low

 

      permeability, but I now would want to caveat that,

 

      I am not sure that is the case.  There is certainly

 

      low systemic availability in general, and that is

 

      probably more the issue, because that makes the in

 

      vivo test plasma levels more difficult to measure.

 

                So, for locally acting drugs, in vivo

 

      dissolution is the key determinant.  So, for the in

 

      vitro dissolution test, we should cover the range

 

                                                               285

 

      of in vivo variables.

 

                So, here is the hypothesis.  If a product

 

      dissolves, two products could be the same

 

      manufacturer, but they just did some reformulation.

 

      If those two products dissolve at the same rate

 

      under all in vivo conditions, such as pH, 6.5, 7,

 

      7.5, maybe a couple more if you want to be really

 

      rigorous.

 

                That means that the two products will

 

      perform the same regardless of what the pH profile

 

      is in an individual subject.  That is what we have

 

      to ensure, and I think we can do that better with

 

      an in vitro test than an in vivo test.

 

                Now, might want to debate do we want to do

 

      6.5, 6.75, you know, but we need to first accept in

 

      principle that a dissolution test is a key crucial,

 

      I would say an essential component of setting a

 

      bioequivalence criteria for a GI drug, because I

 

      think that is the case.

 

                Now, one place we could start is that

 

      biowaivers for Class I drug, if a GI drug is a

 

      Class I drug, high solubility, high permeability,

 

                                                               286

 

      and rapidly dissolving, it is all over, it doesn't

 

      matter.

 

                In this case, the GI drug would be low

 

      permeability or certainly low systemic

 

      availability, it may or may not be low

 

      permeability.  So, I think that is the equivalent

 

      to extending biowaivers to Class III drugs--I am

 

      sorry--that is Class I drugs.

 

                What we are talking about for low

 

      solubility drugs or particularly low permeability

 

      drugs would be extending biowaivers to Class III

 

      drugs, which has been proposed.  I don't know if

 

      there is any examples, and maybe Rob will talk more

 

      about that.

 

                I think for pH, we want to look at

 

      dissolution as a function of pH.  The one product

 

      that I am going to show just some data on, I think

 

      Rob is going to show the same data, so I will be

 

      quick, is mesalamine.  It is in enteric coated

 

      dosage form.

 

                The question would be do we need

 

      surfactants or not, and that would depend on the

 

                                                               287

 

      drug, because if the drug is poorly soluble, then,

 

      we get into the spiro-relevant [ph] dissolution

 

      media, and that is a bigger question.

 

                So, I think we should require a

 

      dissolution test for bioequivalence in the

 

      bioequivalence criteria for acceptance criteria for

 

      GI drugs, that we need to consider the pH and time

 

      that the drug will spend in the stomach and in the

 

      gastrointestinal tract.  I can propose those if you

 

      want to discuss them.

 

                I think what is more important is

 

      accepting or at least advising and recommending to

 

      the FDA that in vitro dissolution testing should be

 

      part of considering the bioequivalence requirement

 

      or testing for a local GI drug.

 

                You would have to use the similarity, you

 

      know, the 10 percent difference or F2 comparison

 

      for dissolution profiles.

 

                The dissolution test actually is a

 

      difficult criteria, I think.  Mesalamine, I will

 

      show just a few slides on that, but mesalamine is

 

      an enteric-coated, local acting drug, and are some

 

                                                               288

 

      dissolution profiles done by Jennifer Dressman, now

 

      at Frankfort, published in European Journal of

 

      Pharmaceutics and Biopharmaceutics, but here are

 

      different products and simulated gastric fluid.

 

                Here is a pH 6.8.  You see, none of the

 

      products would be similar.  They all dissolve at a

 

      different rate, and that is a surprise to

 

      development scientists, and these are different

 

      products.  I don't know if they were approved as

 

      bioequivalent or not actually, but I do not think

 

      they would be bioequivalent in the gastrointestinal

 

      tract even if they were bioequivalent in vivo.

 

                There are some other profiles again

 

      showing that they are quite different.  If you

 

      increase the pH to 7.8, more of them become similar

 

      because they all dissolve rapidly above the pH of

 

      the enteric coating.  So, if you do a dissolution

 

      at a high enough pH, you can make things look

 

      mostly similar, but at the critical pH where

 

      dissolution is occurring, they would be different.

 

                So, I think that the pH dissolution

 

      profile requiring similar dissolution at, let's

 

                                                               289

 

      say, in this case we are looking at I think like a

 

      pH of 6.57, 7.0, 7.5, that pH range.  We can debate

 

      whether you should do pH 6.0 would be critical.

 

                I am going to skip these because these

 

      questions are already up.

 

                So, what I want to propose that the

 

      committee consider and perhaps recommend to the

 

      FDA, I am not sure, I guess we are going to go

 

      through a list of questions that Rob is going to

 

      discuss, but that we do require in vitro

 

      dissolution as part of our bioequivalence testing

 

      for drugs that are locally acting, and that then

 

      the in vivo test, do we need an in vivo test for

 

      safety purposes, for safety assurance purposes, and

 

      do we need a confidence interval test for in vivo.

 

                I think that may be on a drug-by-drug

 

      basis.  You may not want to try and make a decision

 

      for all locally GI drugs.  I think depends on the

 

      pharmacology and metabolism of the drug.

 

                But I can say that if had a rigorous

 

      enough in vitro dissolution test, in vivo testing

 

      would not be required.

 

                                                               290

 

                That's it.  Thank you.

 

                Do you want to have questions now?

 

                DR. KIBBE:  Shall we do that, because I

 

      think Marvin has a question and so do I.  Go ahead.

 

                DR. MEYER:  Real quick, Gordie.

 

                Do you think that F2 is an adequate

 

      parameter to use in making a bioequivalence

 

      decision?

 

                DR. AMIDON:  The answer is I don't know,

 

      Marvin.  I have suggested this to a couple of

 

      people, that we need to evaluate that, and whether

 

      F2 or 50, where did that come from, and is it

 

      enough, and the answer is I don't know.

 

                I think that the statistics of dissolution

 

      and the dissolution variability that you could

 

      allow, that would keep you within the

 

      bioequivalence 80 to 125.  Now, that is not so easy

 

      to answer, and it is going to depend on drug

 

      properties, but I agree, that the F2 or 50 needs

 

      more investigation.

 

                DR. KIBBE:  Anybody else?  Go ahead, Judy.

 

                DR. BOEHLERT:  I finally have a question.

 

                                                               291

 

                By making this suggestion that the in

 

      vitro dissolution is a factor, does that presuppose

 

      that the clinical efficacy of this drug only occurs

 

      in a very narrow pH range, so that this pH

 

      difference you see on dissolution is meaningful,

 

      because they could be clinically equivalent or have

 

      the same action, and have different profiles at

 

      different ph's because where the drug acts is

 

      across the GI tract, and not just one location.

 

                DR. AMIDON:  They could be clinically

 

      equivalent with a different dissolution profile,

 

      but you would have to prove it to me.  No one is

 

      going to do that.

 

                DR. BOEHLERT:  And how would you do that?

 

                DR. AMIDON:  But my answer, I think more

 

      to the point, I think, Judy, is the pH profile

 

      changes through the GI tract, stomach, duodenum,

 

      jejunum, goes 5.5, 6.0, 6.5, and it varies from

 

      subject to subject, and in a fasted/fed state,

 

      during the different Phase I, II, III, of the

 

      fasted/ state.

 

                So, what you want to do is ensure the two

 

                                                               292

 

      products will dissolve under any of the pH

 

      conditions that we would see.  So, if they dissolve

 

      the same, let's say at pH 6.5, 7.0, 7.5, you could

 

      say, well, maybe we should 6.0, maybe 5.5, maybe we

 

      should pretreat for 15 minutes in 10th normal HCL

 

      for a while, pretreat for 2 hours, maybe we should

 

      pretreat at pH 4.0, because that is more like the

 

      average pH in the stomach with food.

 

                So, I think that is more of an issue of

 

      the specific test, and that is going to be a more

 

      elaborate test than our typical so-called

 

      dissolution test.

 

                DR. BOEHLERT:  Actually, I guess my only

 

      concern was if, indeed, the drugs were equivalent

 

      at pH 7.8 where it all dissolved at the same rate,

 

      is it really meaningful that it was different at

 

      lower pH's.

 

                DR. AMIDON:  I would argue that it is,

 

      because the pH in the intestine in humans is more

 

      like around 6.5 to 7.0, and it goes down to 5.0 or

 

      5.5 in the duodenum where you have got the mixing

 

      of gastric acid and pancreatic and bile, so that I

 

                                                               293

 

      would say 7.8 is actually not relevant.

 

                I wouldn't do that.  The highest I would

 

      go would be about 7.5.

 

                DR. KIBBE:  Paul.

 

                DR. FACKLER:  If I can just make a couple

 

      of points.  First, I agree about the dissolution

 

      and how inappropriate the dissolution we do today

 

      is to certainly the way orally absorbed drugs are

 

      taken.  I can't remember the last time I saw

 

      somebody drink 900 milliliters of water with their

 

      tablets.

 

                DR. AMIDON:  Or even 250 ml.

 

                DR. FACKLER:  Or even the 240 or 250 that

 

      we use in the clinics.  But let me ask a question.

 

      For systemic drugs, we look at the plasma

 

      concentrations of the compartment just prior to the

 

      site of effect, and for these drugs, we might look

 

      at the plasma compartment as adjacent to the site

 

      of effect just after it, and so in either case, it

 

      might be a good surrogate measurement for the

 

      amount of drug at the site of action in our

 

      traditional case where we have been doing it for 20

 

                                                               294

 

      years, it is just prior to, but for these drugs, we

 

      might consider it just post, and maybe as a

 

      surrogate marker for the amount of drug that was at

 

      the site of action.

 

                DR. AMIDON:  I would say the following.  I

 

      think that is a good question, Paul, and I thought

 

      some about that.  Two things I would say.  One is

 

      the drug as it spread along the gastrointestinal

 

      tract, depending on how it is released may be

 

      absorbed in different segments, so the drug might

 

      actually be presented to different sites from

 

      different formulations, and still meet a Cmax and

 

      AUC criteria.

 

                Now, if you were to add in Tmax and/or

 

      some absorption rate measurement in comparison,

 

      then, I would agree with you, it would be

 

      equivalent, but that is complicated.  I mean the

 

      FDA has looked into measures of absorption rate

 

      other than Cmax and concluded that there isn't

 

      really any good measurement.

 

                So, the answer is if you measured rate and

 

      Tmax and had criteria on that, I would say then

 

                                                               295

 

      plasma would be just the same basically, but if the

 

      drug is very highly metabolized, so the systemic

 

      availability is low, and you are measuring parent,

 

      and assuming there is no problems with metabolite,

 

      which you can't assume, then, you have got the

 

      variability issues on the plasma site, so the

 

      plasma site could be a much harder test.

 

                DR. KIBBE:  Jurgen.

 

                DR. VENITZ:  I think in general I buy the

 

      idea that in vitro dissolution could predict in

 

      vivo dissolution of those products, but you

 

      mentioned kind of in passing excipient effects on

 

      permeability or metabolism.  What about food

 

      effects?  In other words, food constituents would

 

      considerably at least have the same effects on

 

      either permeability and/or metabolism, which may

 

      affect locally what is going on.

 

                DR. AMIDON:  For bioavailability, I would

 

      agree, Jurgen, it would have a big effect

 

      obviously, but for bioequivalence, I would say the

 

      effects would tend to be washed out by the dilution

 

      of the food of any excipient effect.

 

                                                               296

 

                So, I think when we are comparing two

 

      products, the same drug, I would say it is a less

 

      important issue.

 

                DR. KIBBE:  Anybody have something because

 

      I have got a whole bunch?

 

                DR. MORRIS:  I have just one quick one.

 

      Gordon, were you assuming that this would be done

 

      in the normal dissolution apparatus or is that an

 

      open question?

 

                DR. AMIDON:  That is an open question in

 

      my mind, yes.

 

                DR. MORRIS:  Because I think there is a

 

      fair amount of concern, hydrodynamic at the very

 

      least, with the current apparati.  I just wondered

 

      if you are not restricting it to that.

 

                DR. AMIDON:  I guess my position is

 

      changing dissolution apparatus should be done with

 

      great care and with good justification.

 

                DR. MORRIS:  Oh, absolutely.

 

                DR. AMIDON:  But I believe that a

 

      bioequivalence methodology needs to look at

 

      reflecting in vivo processes, and we should start

 

                                                               297

 

      with that.

 

                DR. MORRIS:  I agree with your premise.

 

                DR. KIBBE:  I follow up on Ken and go down

 

      a road with a lot of variables in it.  I love in

 

      vitro tests if I have control of all the variables,

 

      and when I start losing control of the variables,

 

      then, I start to get worried about the test.

 

                I can imagine two products that have

 

      slightly different excipient compositions who

 

      appear, in a dissolution apparatus, to dissolve

 

      equivalently, but that they aren't presenting the

 

      same amount of drug to the surface of the membrane

 

      for one reason or another.

 

                There might be an excipient in there that

 

      forms cells that trap some of the drug, there might

 

      be an excipient that binds the drug, but in a

 

      dissolution apparatus, the excipient is small

 

      enough so that it goes into solution, but then it

 

      doesn't allow it to release.

 

                I can imagine interactions between food

 

      substances that are different for this dosage form

 

      versus that dosage form.  I mean I really would

 

                                                               298

 

      like to go back to the if you could show me that

 

      both formulations have the same inert or inactive

 

      or excipient ingredients and in the same basic

 

      dosage form, then, I am really happy with

 

      dissolution studies as a mimicker because we are

 

      really looking at the levels of drug in that lumen,

 

      that then gets presented to the surface where it is

 

      supposed to have its effect.

 

                The other thing that I wonder about is if

 

      some of these excipients are permeation enhancers,

 

      than one drug, they both would dissolve the same in

 

      the in vitro dissolution apparatus, but the one

 

      with the permeation enhancer would start to leave

 

      the site where you want it and end up in the blood

 

      supply where you don't want it, and you really have

 

      to link that to some marker at the back end to see

 

      how fast it is leaving where I want it to be.

 

                If one gets into the body in this case

 

      better, that's worse.

 

                DR. AMIDON:  It could be, at least in the

 

      systemic circulation.

 

                DR. KIBBE:  So, the questions that I come

 

                                                               299

 

      back to is what is the dissolution apparatus, what

 

      are the criteria that we have to put on the product

 

      to narrow it down, so that we know that the

 

      dissolution apparatus can tell us if the two are

 

      behaving the same before we put it in a person, and

 

      then when we do that, aren't we better off still

 

      getting minimalist levels in the plasma just to

 

      look for the odd chance that one of them is

 

      permeating better than the other, so that we can

 

      either prevent levels going up, so that we might

 

      have toxicity in one or the other, or prevent

 

      levels being too low, so that we have some

 

      secondary measure.

 

                Now, if we are going to do that, which one

 

      does a better job of actually measuring the drug at

 

      what I would call the biophase where the drug is

 

      having its real action, and I really think they

 

      both measure it incompletely.

 

                DR. AMIDON:  One side and the other.  We

 

      have got it sandwiched in between, right?

 

                DR. KIBBE:  Right.  So, I would argue that

 

      you have to do both somehow.

 

                                                               300

 

                DR. AMIDON:  Maybe we need an intermediate

 

      step here to get more experience.  So, I could see

 

      where we might require some type of pharmacokinetic

 

      measure, plasma levels, as well as dissolution,

 

      both.  That is, I am recommending both.

 

                Now, I believe that when we know enough,

 

      and maybe for some, maybe most, but probably not

 

      all drug products, dissolution would be enough, but

 

      we are not there yet.

 

                DR. KIBBE:  I could support that once I

 

      start narrowing down my variables.

 

                DR. AMIDON:  So, we get into the

 

      discussions of dissolution, but I mean and I could

 

      throw things out, but it is going to take more than

 

      me to kind of evaluate what might be a good

 

      criteria here and dissolution methodology.  I think

 

      what is important at this stage for the committee

 

      is to say, yes, we think this is the right path to

 

      go down and tell the FDA to figure it out, like

 

      Congress does, you know, you go figure out what to

 

      do with bioavailability.

 

                DR. KIBBE:  Ken wants to say something

 

                                                               301

 

      else.

 

                DR. MORRIS:  I have two caveats I guess,

 

      the first of which is that in that dissolution is a

 

      manifestation of the product itself, as we have

 

      discussed, I like that because it is looking at

 

      changes in the product.

 

                But aside from the apparatus issues and

 

      the tactical issues, there is the statistical

 

      sampling issue that we still face with our normal

 

      dissolution testing that I think is probably only

 

      addressed by the consistency that we had hope to

 

      achieve.

 

                DR. AMIDON:  Statistics will be here, too.

 

                DR. MORRIS:  Yes, sir.  I was going to

 

      insult Nozer, but he has left already.

 

                DR. KIBBE:  Marvin.

 

                DR. MEYER:  Gordie, what do you with the

 

      situation where your in vitro is too

 

      discriminating?  You have three different products

 

      and clinically they all work, maybe to different

 

      degrees, but close enough, and yet your dissolution

 

      says there is a 20, 30 percent difference.

 

                                                               302

 

                DR. AMIDON:  Good question, Marvin.  I

 

      have tried when I talk about dissolution and talk

 

      to the dissolution people at the FDA, say do not

 

      use the word discriminating, because that is not

 

      what your job is.  Your job is to ensure in vivo

 

      bioavailability.

 

                If the manufacturer wants to be

 

      discriminating because of his quality standards,

 

      that is fine, the manufacturer can do what he wants

 

      as long as he meets the standards that ensure

 

      bioequivalence.

 

                So, I don't think the FDA should be

 

      regulating on the basis of discriminating

 

      dissolution tests.  I think they should be

 

      regulating on the basis of a test that will ensure

 

      in vivo bioequivalence.  I don't know if anyone

 

      agrees with me, maybe you do, but I don't like the

 

      term discriminating because you can always get

 

      discrimination.

 

                DR. KIBBE:  Ajaz wants to say something.

 

                DR. HUSSAIN:  Gordon, I think you put

 

      something right on, and that is a challenge.  The

 

                                                               303

 

      last several months we have been struggling with

 

      this in a sense.  I did share with you my

 

      presentation to the USP meeting, and so forth.

 

      Since we do not have a good handle on variability

 

      because of the calibration issues, and so forth, I

 

      think the tendency has been at the Agency to say,

 

      all right, how do you minimize that.

 

                I want a big difference, so I feel

 

      confidence, and I think the time has changed to say

 

      what is the intended use, and so forth, so I think

 

      the point you made, I don't think we have consensus

 

      throughout right now, but it is a very important

 

      point, and I think we have to move in that

 

      direction and sort of discuss and debate that.

 

                DR. KOCH:  I just had a quick question

 

      that goes back to some involvement I had maybe over

 

      20 years ago where there was a dog model set up,

 

      and I imagine it could be most animals where they

 

      actually had a trapdoor on his stomach, where you

 

      were able to, in this case, primarily watch

 

      disintegration and subsequent effects by monitoring

 

      plasma levels on dissolution.

 

                                                               304

 

                But is there any development that could be

 

      called pseudo in vivo to check for absorption, not

 

      to do what we are talking about here, but sometimes

 

      there is a question somewhere between the in vitro

 

      and the in vivo?

 

                DR. AMIDON:  I think my answer to that

 

      question is no, there is no way to make a step

 

      forward dosage forms.  I would say we regularly

 

      test in dogs, recognizing, though, that the average

 

      pH in the upper small intestine of dogs is about 1

 

      pH unit higher than humans, so therefore,

 

      enteric-coated products are not going to be

 

      reflected by the dog.

 

                We do controlled release in the dog, but

 

      we only look at the first 6, 8 hours at most,

 

      because of the shorter transit time, and you have

 

      got to just discard any data after that.

 

                The dog, as I understand, is not

 

      basogastric acid secreter, so the pH in the stomach

 

      is much more variable.  So, the answer is I don't

 

      think it will do the job for this, and any smaller

 

      animal you can't do because you can't scale the

 

                                                               305

 

      dosage form down very well, or can't at all maybe,

 

      I guess.  I mean make an enteric-coated tablet for

 

      the rat and scale it up to humans, you have got

 

      bioequivalence issues again.

 

                DR. KIBBE:  The pig is really good.

 

      Meryl?

 

                DR. KAROL:  Am I assuming correctly that

 

      the question really is are we going to test for

 

      bioequivalence locally, that includes the pulmonary

 

      tract, as well as the GI tract?

 

                DR. AMIDON:  Me, no, I am not ready to go

 

      that step in terms of topical or inhalation

 

      although I could make some case that the principles

 

      apply.  The difficulty with topical, for example,

 

      is that you are putting your formulation in direct

 

      contact with the absorbing surface, so the

 

      formulation will affect, let's say, the

 

      permeability of the skin.

 

                The big saving grace about the

 

      gastrointestinal tract is there is this big

 

      dilution in the stomach, and that is why I think

 

      the excipient effects are small.  I mean we have

 

                                                               306

 

      got this big dilution in the stomach, so all the

 

      excipient effects are diluted out.

 

                DR. KIBBE:  We can talk about that.

 

                DR. AMIDON:  Inhalation, also, there is

 

      the physics of the dosage form particle size and

 

      deposition along the GI tract, so I am not an

 

      expert at that, so I am not very knowledgeable

 

      about that.  This is focused on GI.

 

                DR. KIBBE:  Ajaz.

 

                DR. HUSSAIN:  I think the other aspect is

 

      if you recall the discussion Rob Lionberger had

 

      presented on topical skin products, there is a

 

      fundamental principle that is evolving, it is

 

      quality by design.

 

                What that means is in a sense, comparing

 

      formulations at anything worth critical and then

 

      trying to relate that to that, and I think the

 

      aspects of excipient similarity, and so forth, a

 

      lot of this formulation information can be

 

      supportive information that can give you all this,

 

      and so forth.

 

                So, I don't want to discount that in the

 

                                                               307

 

      sense I think Rob Lionberger's presentation on the

 

      topical decision tree had those elements, so in

 

      many ways, I think the proposal for looking at an

 

      in vivo relevant bioequivalence test using in vitro

 

      method is a confirmation of all that similarity or

 

      all the design that sort of comes through.

 

                So, keep that in mind as you think about

 

      it.

 

                DR. KIBBE:  Ajaz, that was clearly the

 

      point we get to.  Once we get an understanding of

 

      the formulation itself, and can look critically at

 

      what is in there besides the active, then, we are

 

      much more confident in what Gordon is proposing as

 

      a way of measuring what is happening because if we

 

      know there is nothing in there that has a habit of

 

      doing the things that might disrupt it, then, we

 

      are done.

 

                I still think that we are going to find

 

      early on it is going to be very comforting, if you

 

      will, to get some blood levels just to make sure

 

      that there is something that we haven't expected

 

      that is happening, but I think we need to go in the

 

                                                               308

 

      direction of dissolution testing.  I think that is

 

      a wonderful way to go in a situation.

 

                Anybody else?

 

                We have another presentation, right?

 

                DR. AMIDON:  Right.  Thank you.

 

                DR. KIBBE:  Thank you.

 

                Dr. Lionberger.

 

                Regulatory Implications and Case Studies

 

                DR. LIONBERGER:  What I am going to talk

 

      about is just give some examples that are

 

      illustrates of how we apply some of these

 

      scientific principles to several different products

 

      with the intention of sort of spurring discussion

 

      although we have already had some very good

 

      discussion.

 

                The first scientific issue is dissolution,

 

      we are not really going to talk about because you

 

      have had a very good discussion about dissolution.

 

                The second scientific issue that I want

 

      you to keep in mind through my presentation is the

 

      issue of sort of how we should interpret

 

      pharmacokinetic measurements that we make on the GI

 

                                                               309

 

      acting drugs.

 

                Certainly, they are related to safety, but

 

      I also want to just indicate that, you know, you

 

      often hear that the peak pharmacokinetics of

 

      locally acting drugs aren't correlated with

 

      therapeutic effect, so I want to sort of focus your

 

      attention on the sort of last point here, on how

 

      the pharmacokinetics of the GI acting drug is

 

      related to formulation performance, and that will

 

      sort of lead into some of the discussion that we

 

      would like to have on how we should use

 

      pharmacokinetic data in evaluating bioequivalence.

 

                So, the first example that I want to talk

 

      about today is for the drug mesalamine.  This is an

 

      anti-inflammatory drug mainly targeting the colon.

 

      It turns out that it is actually pretty rapidly

 

      absorbed from the intestine and this drug can be

 

      measured in the plasma.  There is also some

 

      extensive metabolism pre-systemic circulation, as

 

      well.

 

                The one thing that is sort of interesting

 

      about this drug is sort of a case study, is that

 

                                                               310

 

      there is a wide variety of formulations of this

 

      drug that are currently on the market, so you can

 

      do a lot of sort of comparisons to see which

 

      different types of tests can actually distinguish

 

      between these different formulations.

 

                The sort of key scientific issue that is

 

      driving these formulations is you want the drug to

 

      target basically the colon, but it is rapidly

 

      absorbed, so the formulation technology is either

 

      pro-drugs or some sort of delayed release enteric

 

      coating are designed to sort of keep the drug from

 

      being absorbed until it reaches the target.

 

                So, this sort of raises the issue of

 

      targeting different areas of the gastrointestinal

 

      tract and some of the issues that that might raise.

 

                The first product is sulfasalazine.  This

 

      is the oldest mesalamine product.  It is the third

 

      molecule down in the chemical structures, and it is

 

      a pro-drug that consists of mesalamine moiety and

 

      then the other moiety, sulfapyridine.

 

                For this case, the mesalamine acts

 

      locally.  The other moiety of the product is

 

                                                               311

 

      actually rapidly and quickly absorbed.  So, this

 

      drug is old enough that OGD has actually approved

 

      ANDAs for this product, and the basis for the

 

      bioequivalence determination in this case was

 

      pharmacokinetics, but it was the pharmacokinetics

 

      of the inactive part of the pro-drug, primarily

 

      because it's rapidly absorbed and it has much lower

 

      variability than the active moiety itself.

 

                Also, for this drug, the sulfapyridine

 

      itself has pharmacological activity, and there are

 

      also its systemic exposures is highly related to

 

      some of the safety issues with this product.

 

                So, for this product, this moiety was used

 

      primarily because, as we will see later, there is a

 

      high variability associated with the

 

      pharmacokinetics of the active ingredient itself.

 

                So, that is sort of just one example, and

 

      Lawrence sort of pointed these out, that sort of in

 

      the past, FDA, for these GI acting drugs, has used

 

      sort of a wide variety of different ways to

 

      evaluate bioequivalence, and we would like to sort

 

      of put together a sort of more fundamental

 

                                                               312

 

      scientific framework on sort of when we should use

 

      which aspect.

 

                So, a second mesalamine is the Pentasa

 

      product, and this is a slow release, microgranular

 

      formulation, sort of releases continuously through

 

      the intestine.  It is not really pH dependent on

 

      how it releases.

 

                During some of the development of the

 

      evaluation of the NDA for this product, there are

 

      PK studies attempted for bioequivalence between

 

      pilot and production scale products.  Again, here

 

      the issue was we weren't really able to conclude

 

      bioequivalence because of the high variability of

 

      the active ingredient, but they were able to

 

      establish in vitro/in vivo correlation between

 

      dissolution and the pharmacokinetic studies, and

 

      that was used to demonstrate equivalence between

 

      different pilot and production scale formulations

 

      for that particular product.

 

                So, a third mesalamine formulation is the

 

      Asacol product.  This is a delayed release, coated

 

      formulation, and here, there is pH sensitive

 

                                                               313

 

      dissolution that allows it to target the colon.

 

                So, when you have all of these different

 

      products, we can look at sort of the different

 

      possible ways of testing these products.  The

 

      discussion points that were presented to the

 

      committee were what is the role of dissolution,

 

      pharmacokinetics, clinical studies, so that we can

 

      look at these types of comparative studies between

 

      these different formulations to sort of get at

 

      least some sort of solid basis for discussion.

 

                Gordie in detail showed this data on the

 

      dissolution studies of mesalamine products,

 

      basically different a pH-independent product.

 

      These are mainly European formulations, they are

 

      not the formulations that are marketed in the U.S.

 

                You can definitely see that at the low pH,

 

      you see only the sort of slow release product

 

      dissolving.  As you raise the pH, the different

 

      enterically-coated products start to dissolve

 

      depending on what pH they are particularly

 

      targeting.

 

                So, by choosing appropriate dissolution

 

                                                               314

 

      conditions that are relevant to the in vivo

 

      conditions where the product started, and you can

 

      distinguish between the different products and what

 

      region of the intestine they may be targeting.

 

                There is also some studies that have done

 

      comparative pharmacokinetic studies.  Again, here

 

      is sort of a pellet and tablet type formulation,

 

      not the sort of currently marketed formulations,

 

      but sort of similar ones.

 

                You see in this case, you look at the Cmax

 

      and AUC.  Definitely, in this case, the

 

      pharmacokinetic studies actually show a large

 

      difference between the products, but again, here,

 

      this is just scientific publications for these. It

 

      is small sample sizes, so they didn't evaluate

 

      confidence intervals, but the variability of the

 

      measures for these drugs are very high.

 

                So, that sort of complicates the

 

      determination of bioequivalence using

 

      pharmacokinetic measures.

 

                So, with all these different products on

 

      the market, there has been some interest in trying

 

                                                               315

 

      to say which one clinically is more effective.  So,

 

      there have been, not complete head-to-head trials

 

      between all products, but there have been a large

 

      number of clinical studies, and sort of a recent

 

      review, came to the conclusion that clinical

 

      studies haven't been able to demonstrate

 

      significant differences in the efficacy between

 

      existing mesalamine formulations and stuff, two

 

      examples here, but there are sort of many studies

 

      available in the literature.

 

                So, if you were trying to sort of

 

      determine equivalence between two formulations, you

 

      might have a very hard time using the clinical

 

      study to have a sensitive discrimination of

 

      formulation performance, because these existing

 

      formulations, which use very different

 

      technologies, aren't very well distinguished by

 

      sort of clinical studies using the usual efficacy

 

      endpoints.

 

                So, if we just summarize the mesalamine

 

      example, if we want to sort of distinguish the

 

      current products, we would probably yes, if we

 

                                                               316

 

      chose the right dissolution criteria, we could

 

      clearly see the difference between the products.

 

                Pharmacokinetics, it looks like again we

 

      could see the difference especially because if the

 

      products release early, they are rapidly absorbed.

 

      If they are delayed into the colon, then, the

 

      absorption is much slower, so differences in local

 

      release actually do show up in the pharmacokinetics

 

      through especially Cmax for this case.

 

                But again, with this particular product,

 

      there is the issue of pharmacokinetics of highly

 

      variables.  If you want to do clinical comparisons

 

      in terms of the bioequivalence study, again, that

 

      would be very challenging in terms of getting a

 

      sensitive test of the formulation differences.

 

                So, just to bring in a slightly different

 

      example, another example of a locally acting

 

      product is Acarbose. This is an intestinal enzyme

 

      inhibitor that acts to reduce glucose absorption.

 

      For this product, there is no measurable

 

      absorption, so you can't use pharmacokinetic

 

      studies, however, there are pharmacodynamic

 

                                                               317

 

      endpoints available.

 

                Again, you can look at changes in glucose

 

      or insulin level in response to a meal with the use

 

      of this drug or a comparator product.

 

                Here, one sort of interesting thing to

 

      think about is when we think about the

 

      pharmacokinetic studies, like the pharmacodynamic

 

      endpoints, are usually downstream measures of the

 

      formulation performance, in the same way that the

 

      pharmacokinetic measurements are here.  So, there

 

      is some sort of mathematical similarity between how

 

      we might interpret pharmacodynamic endpoints and

 

      pharmacokinetic measurements for GI acting

 

      products.

 

                Another example of a product where there

 

      is not much detectable absorption is

 

      cholestyramine.  This is a bile acid sequestrant,

 

      essentially binds to cholesterol in the intestine.

 

                This is sort of an older product.  In

 

      1993, FDA guidance recommended using an in vitro

 

      binding assay to demonstrate equivalence of these

 

      products, so there is no dissolution of

 

                                                               318

 

      pharmacokinetics.  These assays measured affinity

 

      and capacity.

 

                One of the issues, here, we talked a

 

      little about the role of excipients.  For these

 

      types of products that are involved in binding

 

      there, then, you sort of worry that if there is

 

      differences in the excipients in the formulation,

 

      that that might make a difference in how they bind

 

      to other products, and these types of in vitro

 

      binding assays can be valuable and interesting,

 

      those types of concerns if they are relevant to a

 

      particular product.

 

                So, before I lead into our discussion, I

 

      just want to sort of return again to some of the

 

      scientific issues that were raised by the GI acting

 

      drugs.

 

                The first is the BCS classifications and

 

      biowaivers again for systemic drugs.  If we have

 

      high permeability and high solubility drugs in

 

      rapidly dissolving dosage forms, we often consider

 

      waiving in vivo bioequivalence studies.

 

                So, the question is how should we extend

 

                                                               319

 

      this to GI acting drugs.  I think one more question

 

      is what should the permeability play.  If

 

      permeability doesn't play any role in the

 

      absorption process, should we extend the biowaivers

 

      to sort of all highly soluble drugs in rapidly

 

      dissolving forms irrespective of what their

 

      permeability is, or is there something about the

 

      classification of drugs in terms of high

 

      permeability, low permeability that may make that

 

      more risky, perhaps interactions with excipients or

 

      the role of absorption in the intestinal tract.

 

                So, that is one of the issues that we

 

      might like to have some discussion on.

 

                Again, I just want to come back to the

 

      issue of the role of pharmacokinetic studies in the

 

      absorption from the GI tract.  Again, for a

 

      systemic drug, what you see is that the formulation

 

      performance is what we are really trying to make a

 

      determination about.  Again, this essentially is

 

      controlled by the dissolution.

 

                The drug goes through the absorption

 

      process, reaches the plasma concentration, and that

 

                                                               320

 

      is the place where you take a pharmacokinetic

 

      sample, and that is also the place where the effect

 

      of the drug takes place.

 

                So, the difference for a GI acting drug is

 

      essentially the relationship between sort of the

 

      formulation performance, pharmacokinetic sample,

 

      that is still the same, but the only thing that

 

      sort of moved is where the effect is taking place.

 

                If you think about how we conduct

 

      bioequivalence studies, we usually conduct them in

 

      healthy people.  We don't really concern ourselves

 

      with the effect, so that whatever connection we are

 

      making for systemic drugs between PK sampling and

 

      formulation performance, the connection is still

 

      there for the GI acting drugs.

 

                So, I think the big concern with looking

 

      at the pharmacokinetic studies in the GI acting

 

      drugs is essentially when, for the systemic drugs,

 

      since we know the effect is taking place where we

 

      are taking the sampling, we have some sort of

 

      intrinsic way to know what a significant difference

 

      is.

 

                                                               321

 

                We sort of say 80 to 125 percent

 

      difference in the plasma concentration is sort of a

 

      general definition of clinical significance, so we

 

      know that if we meet that, that sort of gives us an

 

      idea of what equivalent formulation performance is.

 

                When we go to the GI acting case, where

 

      the effect is now separate, we don't have that

 

      connection as to what the calibration is between

 

      difference in pharmacokinetic sampling and a

 

      significance clinical effect in the same way that

 

      we do for the systemic drugs, so that I think is

 

      the issue for interpreting the pharmacokinetic

 

      studies is we don't have the sort of intrinsic

 

      calibration of how significant the effect on

 

      formulation performance is.

 

                But still the relationship between

 

      formulation performance and pharmacokinetics is

 

      still there in both cases in a way that maybe it is

 

      not for other locally acting drugs, say, inhalation

 

      products where you have the case where if the

 

      product reaches the lungs, it also can be absorbed

 

      by different pathways, and you are not sure that it

 

                                                               322

 

      is passing through the exact same pathway.

 

                For the GI acting drugs, you know that in

 

      both cases, it is being absorbed through the same

 

      site.

 

                Just to lead into sort of the specific

 

      questions that we wanted you to discuss, just sort

 

      of outline, a sort of potential framework for

 

      thinking about bioequivalence of locally acting

 

      drugs, sort of the first point would be again the

 

      sort of critical importance of dissolution testing

 

      in conditions based on understanding of the

 

      formulation and how it interacts with the in vivo

 

      environment, so choosing the right conditions for

 

      in vivo and in vitro dissolution testing.

 

                The second part of that is pharmacokinetic

 

      and pharmacodynamic studies.  Again, the sort of

 

      potential we always see for those is really, they

 

      sort of confirm the dissolution testing.  They

 

      confirm the relationship between the in vitro

 

      dissolution testing and the in vivo dissolution,

 

      which determines product behavior.

 

                They are also important for assessing

 

                                                               323

 

      systemic exposure in terms of any type of safety

 

      concern.

 

                The third element of the framework is

 

      concern about excipient interactions, and if, say,

 

      the product's mechanism of action is binding to

 

      like the cholestyramine mean example, binding to

 

      something in the GI tract where an excipient could

 

      be competitive or inhibitory for that binding

 

      process, it might be useful to require some sort of

 

      in vitro assay for that type of process for

 

      particular products if there is a mechanistic

 

      reason why the excipient interaction may be

 

      important.

 

                So, here, I just want to remind you of the

 

      discussion questions that we suggested to you.

 

                We wanted your input on the role of

 

      pharmacokinetic studies, the role of the in vitro

 

      dissolution tests, and the role of clinical studies

 

      in these particular products.

 

                I have sort of listed out sort of slightly

 

      more detailed versions of these questions, you

 

      know, how should we use the pharmacokinetics data,

 

                                                               324

 

      if it's measurable, to evaluate formulation

 

      performance?  What drug specific information would

 

      be valuable in sort of calibrating our

 

      interpretation of pharmacokinetic studies, when it

 

      would be valuable, when it would not be valuable?

 

                When is it possible to use dissolution

 

      testing alone to demonstrate bioequivalence, and

 

      when do we actually need the confirmatory data from

 

      pharmacokinetic or pharmacodynamic studies, as

 

      well?

 

                When should comparative clinical trials be

 

      conducted, what types of issues there?

 

                The final question on who we should look

 

      at extending the BSC-based biowaivers for GI acting

 

      drugs.

 

                With that, hopefully, we will be able to

 

      have some more discussion on some of these issues.

 

                Committee Discussion and Recommendations

 

                DR. KIBBE:  Marvin is ready.

 

                DR. MEYER:  Kind of relating to your in

 

      vitro question, are there any drug products that

 

      are known to act locally in an undissolved state,

 

                                                               325

 

      particles, fine particles?

 

                DR. KIBBE:  What about Sucralfate?

 

                DR. MEYER:  Yes, Sucralfate.  Dissolution,

 

      PK, would that work?

 

                DR. LIONBERGER:  I think you have to

 

      consider the mechanism of action.  I am thinking

 

      primarily here of drugs,  you know, where the sort

 

      of mechanism of action is distinct from the

 

      formulation.  I think when you have products where

 

      the sort of mechanism of action is very connected

 

      to how the drug is formulated, then, you have to

 

      have some measure of the formulation performance in

 

      vivo.

 

                So, if you were thinking mainly of drugs

 

      where the drug is released from a formulation

 

      before it reaches the site of action, and I think

 

      there are sort of other issues where the

 

      formulation acts, just the formulation or, you

 

      know, manufacturing acts directly.

 

                I think that issue probably will come up a

 

      lot when we look at nanotechnology.

 

                DR. MEYER:  I sort of subscribe to

 

                                                               326

 

      Gordie's point of view.  There is probably a

 

      dissolution test that will work, and I think we

 

      know enough about dissolution testing if we want to

 

      use paddle and basket and three different RPMs and

 

      five different pH's, and surfactants, and

 

      everything that anyone has ever done, and two

 

      products are equivalent under a myriad of

 

      conditions, they are probably going to be

 

      bioequivalent.  "Probably" is not a good regulatory

 

      word.

 

                DR. AMIDON:  It would be very low risk of

 

      bioINequivalence.

 

                DR. MEYER:  Somebody would argue, well,

 

      that's overkill, you shouldn't have to do 89.

 

      Well, that is a lot cheaper than doing a clinical

 

      trial with these products, so that might be one

 

      approach.

 

                DR. KIBBE:  I think Paul would agree that

 

      it is cheaper than doing a clinical trial.

 

                DR. FACKLER:  Could I make a couple

 

      comments?

 

                DR. KIBBE:  Oh, please.

 

                                                               327

 

                DR. FACKLER:  Just to answer your

 

      question, there are some rectal suspensions that

 

      might fall into the category of drugs that are

 

      effective without dissolving.

 

                My understanding of the lower part of the

 

      colon is that it is relatively water-free, and

 

      these undissolved-- mesalamine being one of them,

 

      by the way--products, hydrocortisone is another

 

      one, both of which, by the way, OGD has approved on

 

      the basis of pharmacokinetic comparability.

 

                The other way that mesalamine, for what it

 

      is worth, is delivered to the colon is by

 

      suppository in the United States, I think.  So,

 

      there is an oral tablet, there is an oral capsule,

 

      there is a rectal suspension, and a suppository,

 

      all of which are equally efficacious and making you

 

      wonder about the utility of clinical studies.  I

 

      will just leave it at that.

 

                DR. KIBBE:  That is a valid point.  Once

 

      you have lots of different routes of

 

      administration, and for a local effect, and the

 

      formulation effects clearly go away when you look

 

                                                               328

 

      at the clinical impact, but remember that clinical

 

      endpoints are very wide goalposts, and we tend to,

 

      I guess being slightly anal-retentive, want

 

      narrower ones for regulatory purposes.

 

                Just a small point.  Most drugs don't act

 

      in the central blood supply, they act someplace

 

      else, and even though we measure, we measure

 

      central because we assume, having never actually

 

      verified this, assume that they are in relative

 

      strict proportionality to the amount of molecules

 

      of drug at the actual biophase, and that is the

 

      whole basis for kinetics and what Marvin and I have

 

      done for all our lives, so we are reticent to give

 

      that up, but you can't say that that is where the

 

      drug works, because that is not where it is working

 

      either.

 

                DR. KIBBE:  Ken.

 

                DR. MORRIS:  I just had a question because

 

      I think, Art, actually, you mentioned Sucralfate.

 

      What is the criteria for bioequivalence?  That is

 

      not absorbed at all, right?

 

                DR. KIBBE:  Right.

 

                                                               329

 

                DR. MORRIS:  So, what is the criteria for

 

      bioequivalence for that?

 

                DR. AMIDON:  I know originally, they were

 

      doing clinical studies.  There were clinical

 

      comparison studies.

 

                DR. MORRIS:  It just seemed to me I mean

 

      it would make no sense to do pharmacokinetic

 

      studies on cholestyramine, which never dissolves

 

      even when it is active.

 

                DR. KOCH:  Just to add to that, the

 

      cholestyramine is an interesting one, because from

 

      an in vitro type, you can't really duplicate the

 

      sequestering.  I mean bile acid is just one of the

 

      things that it sequesters it.  Basically, it's a

 

      handful of ion exchange resin, and ion exchange

 

      resins are trained to go after a lot of things

 

      where it can pick up that ion.

 

                So, that would be a difficult one I think

 

      to just run one simple in vitro test.

 

                Another point that I thought of when we

 

      talked about the suppository, Ajaz, when you go to

 

      Europe, do you do a check in terms of dosage forms,

 

                                                               330

 

      as well, because it was very interesting on a

 

      European assignment, it turns out that more of our

 

      drug deliveries were suppositories than they were

 

      tablets, and someday we will start to see that as

 

      another way of administration, particularly as

 

      dosages get smaller and smaller.

 

                DR. KIBBE:  I think suppositories are much

 

      better accepted among the populace in France and

 

      Germany than they are here.

 

                You had a comment.

 

                DR. AMIDON:  I worked with the FDA

 

      extensively and did a lot of in vitro testing on

 

      the bile acid resins, with different bile acids

 

      under differing conditions, and so it is a fairly

 

      rigorous test in terms of the capacity of the ion

 

      exchange resins to bind relevant bile salts.

 

                I think if you look at the guidance, you

 

      would look at it and say if two resins appear the

 

      same under all of these conditions, they are

 

      likely, likely, the risk of bioINequivalence is

 

      low.

 

                Of course, the questions in plasma, what

 

                                                               331

 

      are you going to measure?  You wanted to do

 

      cholesterol lowering.  That is a long, extensive

 

      study, so clinical studies, at least I interpret

 

      here, clinical studies meaning efficacy studies are

 

      much more complicated, much more variable, and I

 

      think quite insensitive to formulation differences.

 

                So, I think we can make an adequate case,

 

      and if you come up with something that you think

 

      might affect the in vivo performance, we can

 

      enumerate what happens in all of the components in

 

      the GI tract, and they can be tested, so we can do

 

      an in vitro test to see if it has an effect and

 

      decide whether it is relevant or not.

 

                DR. YU:  I just wanted to comment on

 

      Paul's comments for rectal suspensions, especially

 

      for the mesalamines, when we look at the

 

      pharmacokinetics, we also look at a dissolution

 

      very closely.  Thank you.

 

                DR. KIBBE:  So, you have a whole body of

 

      data then on pre-existing products of varying

 

      formulation, so the Agency actually has a real good

 

      handle on whether or not there are a diversity of

 

                                                               332

 

      excipients and could actually do dissolution

 

      testing on samples of all the products already on

 

      the market in various environments to come up with

 

      a criteria.

 

                DR. YU:  That's correct.

 

                DR. KIBBE:  Anybody else got anything?

 

                Ajaz has a comment.  Good.

 

                DR. HUSSAIN:  As I think about these

 

      questions, I think Dr. Amidon and Ken Morris both

 

      have pointed out in a sense I think what is in

 

      vitro test conditions and how appropriate they are.

 

      That is a significant challenge.

 

                I don't want to sort of jump in and say

 

      all right, BCS Class I was highly soluble, highly

 

      permeable, 900 ml, and so forth, because the volume

 

      and the hydrodynamics, and so forth, I think we

 

      have to give some thought to how we would approach

 

      that, so it is not a trivial matter.

 

                DR. KIBBE:  Go ahead, Ken.

 

                DR. MORRIS:  I guess I would just echo

 

      that.  The principle I think is sound, you know,

 

      which is no surprise coming from Gordon, but the

 

                                                               333

 

      tactical aspects of that really are quite a

 

      challenge.  There is still a lot of work to do in

 

      terms of dissolution testing.  As Gordon said,

 

      redesigning the dissolution test is no seed for the

 

      faint hearted, I mean that is something that is

 

      going to really take some serious scientific and

 

      engineering work.

 

                DR. MEYER:  Is it correct, Gordon, that a

 

      Class I BCS is likely to appear, at least to some

 

      extent, systemically, and a Class III similarly,

 

      maybe not so much so, but still, because of high

 

      solubility, you are likely to have something you

 

      can measure, and therefore, you could do a PK

 

      study?

 

                DR. VENITZ:  If it has high first pass

 

      effect, it might not be systemic.

 

                DR. MEYER:  That's true.

 

                DR. KIBBE:  Detectable levels are going to

 

      be a problem.

 

                DR. AMIDON:  Class III drugs tend to be

 

      not very highly metabolized, so it would probably

 

      work there, and that is where I think it is the

 

                                                               334

 

      most important, because for a low permeability

 

      drug, there is obviously permeability dependence

 

      along the GI tract, because there is some

 

      absorption, and then it stops.  It has to because

 

      it is not fully absorbed.  So, I think Class III

 

      drugs is where it is more critical.

 

                DR. MEYER:  Therefore, you wouldn't need

 

      to give a waiver,  you could do PK.

 

                DR. AMIDON:  You could.  I am not saying

 

      you can't do PK.  In fact, PK plasma levels in

 

      general, even for GI drugs, I mean if you can

 

      measure something, you know, it would give you the

 

      highest assurance.  The question is what is the

 

      best test, and broadly, for bioequivalence, Cmax,

 

      AUC is our gold standard.  I think our focus on

 

      Cmax, AUC has kind of preempted us from thinking

 

      about what is the real issues here, which are for

 

      oral absorption and/or, for GI, the GI locally

 

      acting drugs, the dissolution process is where the

 

      action is at, and when we want to set standards,

 

      some drugs are going to be simple and some drugs

 

      are going to be complicated, so let's try and

 

                                                               335

 

      decide where we can simplify the standard and make

 

      it maker, and then where it is complicated, well,

 

      that is where science is today.

 

                DR. KIBBE:  I agree with Gordon.  I think

 

      if you have a lot of background data, we can safely

 

      go to a dissolution test with something like this.

 

      In the absence of it, it is always nice to have a

 

      little bit of PK data, blood level data, maybe a

 

      simplified study just to get a sense for the

 

      levels, because we want to be careful of toxicity

 

      and equivalence, and it is going to be case by

 

      case.

 

                Anybody else?  Jurgen.

 

                DR. VENITZ:  I was just going to speak and

 

      for being a former clinician, in favor of clinical

 

      studies.  I mean everybody here is mentioning a

 

      true statement.  They are not very sensitive to

 

      formulation effects, but on the other hand, they

 

      are the ultimate relevant test.  I mean they make

 

      what we are doing clinically relevant.

 

                So, as much as I am personally in favor

 

      and moving along with looking at dissolution

 

                                                               336

 

      testing as the base of your surrogate of in vivo

 

      bioequivalence, there is a price to be paid, and

 

      that is, we are going to find differences in those

 

      dissolution tests between formulations that

 

      clinically are irrelevant.

 

                So, we are looking for discriminating

 

      tests--excuse the term--that discriminates between

 

      formulation differences that are clinically

 

      probably meaningless.

 

                DR. KIBBE:  And the question I guess boils

 

      down to an economic one, do I want to spend the

 

      money to do a clinical test to show obviously that

 

      the differences are meaningless, or can I do a

 

      fairly well designed dissolution test which doesn't

 

      cost me much and is very discriminating, and if I

 

      pass that, I am guaranteed that I will be okay on

 

      the clinic, and that is really what these tests

 

      are.  These are surrogates for the ultimate use of

 

      the drug in 400,000 people.

 

                DR. VENITZ:  We had a similar discussion a

 

      couple of years ago when we talked about intranasal

 

      products, and I guess this committee voted in

 

                                                               337

 

      favor, and the FDA ultimately accepted the fact

 

      that the only way to assess bioequivalence of

 

      intranasal products is the walk in the park, in

 

      other words, a clinical test.

 

                Given the fact from my perspective that I

 

      think we understand much more about GI dissolution,

 

      GI absorption, all of which you presented, Gordon,

 

      I am personally comfortable in moving along with

 

      that, and not making the clinical gold standard a

 

      requirement, but I am just cautioning that in the

 

      process, you are going to throw out formulations

 

      that are clinically probably equivalent.

 

                DR. KIBBE:  To ahead, Ajaz.

 

                DR. HUSSAIN:  That is one of the reasons,

 

      I think, why we pushed the concept of quality by

 

      design, and so forth, because all the relevant

 

      formulation information and all that has never been

 

      brought into that discussion.

 

                It was simply a test to test comparison

 

      discussion, so over the last  several years, we

 

      have brought that discussion up, and then as you go

 

      towards understanding your formulation,

 

                                                               338

 

      understanding what pharmaceutical equivalence could

 

      mean from that perspective, we actually can open

 

      that debate again because of that.

 

                DR. KIBBE:  Ken.

 

                DR. MORRIS:  Just a quick question,

 

      Jurgen.  You are still doing dose-ranging studies

 

      when you are doing the initial development, I

 

      guess, so if you are going to use the prior

 

      knowledge, use the dose-ranging studies, doesn't

 

      that help you when it comes time to determine

 

      whether or not the tests are over-discriminating or

 

      not?

 

                DR. VENITZ:  But I mean most of the time,

 

      even a two-fold dose range, you may not be able to

 

      distinguish clinically, so you are talking about

 

      100 percent difference in formulation performance,

 

      and clinically, you may not be able to tell the

 

      difference.

 

                DR. KIBBE:  Paul has another one.

 

                DR. FACKLER:  I just want to correct

 

      something.  On the nasal products, the clinical

 

      study is required, but in addition to that, the

 

                                                               339

 

      only way to get a generic product approved is to

 

      also pass a PK study and also pass in vitro plume

 

      geometry and spray pattern.

 

                So, one of those three isn't good enough,

 

      two of those three aren't good enough, all three

 

      need to pass in order for the nasal products,

 

      which, in my personal opinion, is overkill for

 

      demonstrating that the two products are equivalent.

 

      The clinical study alone should have been enough.

 

      If the patients are being benefited equally, the

 

      products to some extent are bioequivalent.

 

                The other thing I wanted to correct was

 

      just that the variability of mesalamine is

 

      admittedly high, but not so high that it can't be

 

      dealt with in a pharmacokinetic sense.

 

                DR. YU:  At the last Advisory Committee

 

      meeting, we had a topic on how to deal with highly

 

      variable.

 

                DR. FACKLER:  Only that I know there are

 

      some relatively new data on some mesalamine

 

      products available to the Agency, so that the

 

      variability, at least from rectal suspensions, is

 

                                                               340

 

      defined and was manageable in an BE sense.

 

                DR. VENITZ:  Just to follow up on that,

 

      you are correct.  I mean for the intranasally

 

      administered drugs, they have to pass all three.

 

      What I would like to see for this in terms of the

 

      future progress, would be not to get to that level.

 

      If we accept in vitro dissolution as a surrogate of

 

      in vivo dissolution, as a surrogate of in vivo

 

      bioequivalence, let's stick with it.

 

                If you decide that we don't

 

      mechanistically understand enough what is going on

 

      and we require clinical study, let's stick with the

 

      clinical study.

 

                DR. KIBBE:  Thank you, Jurgen.

 

                Anybody else?  I see by the clock on the

 

      wall that we are running out of time.  Have we

 

      given you enough guidance on this one to move

 

      forward without taking any formal votes?  Lawrence

 

      wants some more information.

 

                DR. YU:  That's correct.  Thank you.

 

                DR. KIBBE:  He wants to thank me.  That's

 

      good.

 

                                                               341

 

                I have on my calendar of events that there

 

      is a summary and conclusion, summary remarks, and I

 

      have two names, and they are looking at each other

 

      like which one of you is going to say anything.  I

 

      would be happy to just rule you of order and close,

 

      if you don't have anything to say.

 

                Go ahead.

 

                     Conclusion and Summary Remarks

 

                DR. HUSSAIN:  I think again as the

 

      previous meeting, I think the discussions were very

 

      valuable and I think help us think more.  The one I

 

      think you probably for the first time got an

 

      opportunity to see the range of laboratory and

 

      other such activities that we have ongoing, and I

 

      think the Critical Path Initiative clearly is not

 

      just lab based, it is much broader than that, but

 

      that discussion allowed us to think more carefully

 

      about how to approach the Critical Path Initiative.

 

                It also helped us to start thinking about

 

      how do you align such programs, especially the

 

      laboratory programs, to be targeted and the key

 

      questions.  The challenge is great and I think we

 

                                                               342

 

      want to maintain as many best practices as we have,

 

      and you did see a number of best practices sort of

 

      come out in the discussion, and maintain that, and

 

      bring all the offices in OPS to be aligned

 

      together.

 

                The immediate office project that we

 

      articulated, the three projects, all interrelated,

 

      I think will be a means to not only identify the

 

      best practices, but also to bring a system approach

 

      to address uncertainty and complexity, and I think

 

      that would be the key aspect and in many ways, that

 

      allows us to approach bioequivalence, follow-on

 

      proteins, generic drugs, all of those challenges

 

      next year in a systematic manner.

 

                So, I think in a number of cases, I think

 

      irrespective of what that pathway for these

 

      products might be, the follow-on proteins, the

 

      scientific framework for the decisionmaking process

 

      should be common irrespective of that, and it

 

      should be related to the uncertainty and complexity

 

      of the dosage form set of products that we have.

 

                At the same time, I think you saw an

 

                                                               343

 

      impressive array of laboratory research from

 

      biology to quality, and how do we align that, I

 

      think is a significant challenge.  We have sort of

 

      summarized that discussion early this morning, and

 

      the key questions, the metrics, I think will be the

 

      key part for sort of making sure whatever approach

 

      we use is measurable and then quantifiable in terms

 

      of its benefit to the critical path, and so forth.

 

                But I do want to emphasize in the sense

 

      that FDA is only one part of that critical path.

 

      Industry, academia, and other agencies play an

 

      equally important role.  Our role will be more also

 

      of coordination, but the need for research,

 

      especially fundamental research in this area and

 

      need for public funding is acute.

 

                I do want to go back and say the

 

      formulation development, manufacturing has been a

 

      neglected area, especially in the U.S., and if you

 

      don't bring the focus on that, I think we are

 

      already 10 years behind Europe and Japan in many of

 

      these areas, so we will lost that part of the

 

      industry, so that is important, so seek your help

 

                                                               344

 

      to make that case also.

 

                I think in terms of the gaps going to the

 

      desired state, Helen outlined some of the key

 

      fundamental organizational gaps.  Some

 

      reorganization is already occurring.  White Oak

 

      provides an opportunity to really bring a team

 

      approach and peer review process to the CMC

 

      function in Office of New Drug Chemistry, but at

 

      the same time, I think one key aspect is a

 

      question-based CMC review process which focuses on

 

      risk.

 

                That is already in the works, but also

 

      support that with tools that we have not often

 

      utilized in this arena, and that is chemometrics

 

      modeling and other aspects of that.

 

                I have a virtual team for chemometrics

 

      right now, but I think we are adding people with

 

      computational fluid dynamics, and others, to really

 

      make a core team that will support the review

 

      function in many aspects.

 

                For example, computational fluid dynamics

 

      is the issue of hydrodynamics, issue of inhalation

 

                                                               345

 

      products, and so forth.  So, hopefully, we will

 

      have that team up and running soon.

 

                I think the science gaps are significant

 

      from a training perspective, but those are not, in

 

      my opinion, unsurmountable.  I think we have seen,

 

      we have the expertise within the Office of New Drug

 

      Chemistry, Generic Drugs, and so forth.  It is

 

      simply identifying and aligning that expertise bear

 

      on some of those challenges, but also provide a

 

      training program for all of our reviewers.

 

                In fact, I really think the PAT training

 

      program opened up a lot of opportunities for our

 

      staff to excel in areas and become leaders

 

      worldwide, and although we cannot do the entire

 

      period, training for all of our staff immediately,

 

      especially the practicum part of it, but as we go

 

      to the next PAT training program, we intend to open

 

      the didactic sessions that we have locally to all

 

      of CMC reviewers to be part of it.  So, we will

 

      bring that onboard.

 

                I think Jon outlined for you some of the

 

      directions we will make more in policy.  Jon is

 

                                                               346

 

      aggressively moving in that direction and I think

 

      his leadership will help us align.  A number of

 

      people have joined his group.  He has a group that

 

      is focused on that now.

 

                So, policy alignment and the OPS

 

      Coordinating Committee with Gary Buehler and Keith

 

      Webber co-chairing that, sort of brings all in one

 

      place now to sort of make sure that the policies

 

      that evolve are aligned with where we want to go.

 

                The issue I think I do want to mention,

 

      the issue of two-tiered approach, I think there is

 

      a risk of that, clearly, there is a risk of that,

 

      but I think we will try at least at the draft 3.1

 

      for Q8, at least I felt that the language was

 

      written not to invoke a two-tiered approach. It's a

 

      continuum, it will be a challenge to manage, but I

 

      think we will get there.

 

                I am hoping in Yokohama, Japan, starting

 

      November 12th, we will bring Q8 to Step 2.  I am

 

      keeping my fingers crossed.  As soon as that

 

      happens, I think things will start moving rather

 

      quickly.

 

                                                               347

 

                Pharmaceutical development information is

 

      already coming in.  Many companies were willing to

 

      take the first step, and have done so, and the

 

      initial experience is positive, but we will have a

 

      quality system, along with peer review process, to

 

      make sure consistency and proper utilization of

 

      that comes in.

 

                I thin, the other two topics are probably

 

      fresh in our minds.  I think bioINequivalence is a

 

      significant challenge, but it is a challenge right

 

      now we are facing to minimize our resources being

 

      spent in things which we think are not value added,

 

      and I think as we move forward, the discussion here

 

      will be helpful.

 

                We will probably not bring that topic back

 

      and we will probably come with an approach, and

 

      then solve that in a way which is consistent with

 

      the way we do it, so I think the discussion will be

 

      helpful, but I still feel, I think Jurgan and

 

      Professor Nozer Singpurwalla, I think we have to

 

      start using prior information, prior knowledge more

 

      effectively, and especially with biostudies that we

 

                                                               348

 

      will have access to, I think it allows us to be

 

      more proactive and make decisions more quickly, so

 

      that Gary and his staff really don't have to spend

 

      so much time in answering these questions in a

 

      legal perspective, and so forth.

 

                Locally acting products clearly are part

 

      of the critical path for the generic drugs.  It is

 

      not only GI, inhalation, topical, it is an entire

 

      area of research that Lawrence will have to sort of

 

      spearhead and move forward, and that is a critical

 

      path research for generic drugs.

 

                Approval of generic drugs in a timely

 

      manner hinges upon that.  I think that PAT concept,

 

      the cGMP, the Quality by Design are all positioned

 

      right to help generics and help innovators all

 

      together, and so you will see that happen.

 

                With dissolution testing, I do want to

 

      sort of say that I think dissolution testing, we

 

      have to think carefully about the variability

 

      aspects of that and how we calibrate. In many ways,

 

      I think our labs have started putting a document

 

      together.  They feel that mechanical calibrators

 

                                                               349

 

      and others are sufficient and relying on an

 

      external calibrator of poor quality actually is

 

      diverting attention away and actually creating

 

      problems, unnecessary problems, so we will issue

 

      something on that soon hopefully.

 

                With that, I will hand it over to Helen.

 

                MS. WINKLE:  Ajaz did a wonderful job of

 

      recognizing, I think, the contributions that the

 

      committee made.  I think there were some excellent

 

      discussion over the last two days and some

 

      excellent recommendations that have been made to us

 

      on things that we need to focus on more and areas

 

      that we need to do more planning and even more

 

      research.

 

                I did also want to mention I thought that

 

      the presentations made by Dr. Boehlert and Dr.

 

      O'Neill on the two workings groups, the

 

      subcommittee for Dr. Boehlert and the working group

 

      for Dr. O'Neill, were very beneficial to those

 

      discussions.  I think both groups are working hard

 

      to accomplish a lot and to get answers back to us

 

      that are really necessary.

 

                                                               350

 

                The Manufacturing Subcommittee at the last

 

      meeting I thought did an excellent job, and think

 

      their report back to you yesterday was indicative

 

      of how much effort they are putting in to helping

 

      make some of the recommendations that we need to

 

      move forward.

 

                Also, with the Dose Uniformity Working

 

      Group, they, too, have worked very hard during the

 

      year, and I think that the report Bob made was well

 

      accepted by the committee and is a good indication

 

      of how these working groups, too, can be beneficial

 

      to the committee in making recommendations to us in

 

      the future.

 

                I do have some other little things,

 

      though, I want to talk about, and that is the two

 

      people that are leaving the committee.  It is a sad

 

      time for us, I think here at FDA, because we have

 

      appreciated both Marv and Art's contributions.

 

      They have been very, very significant in helping

 

      direct us at the Agency in the directions that we

 

      really need to go, and they have also provided a

 

      great deal of scientific expertise and knowledge,

 

                                                               351

 

      not only on the committee, but in other aspects,

 

      too, and they have been very valuable to us.

 

                The one thing, too, I would like to add is

 

      they have also added a great deal of humor to this

 

      committee, which I think many of us are going to

 

      miss.  Now, how long we will miss it is

 

      questionable, because they are both SGEs and can be

 

      called back at anytime--just like Gordon is over

 

      there shaking his head--they can be called back at

 

      anytime to participate in different discussions,

 

      and I think that we are probably going to continue

 

      to take advantage of them.

 

                But today, being their last day on the

 

      committee, I do want to present them with these

 

      plaques in recognition of their service to the

 

      Advisory Committee.

 

                The first one is to Dr. Kibbe.  Not only

 

      has he been an excellent, excellent member of the

 

      committee, he has also been a very, very

 

      thought-provoking chair even though a little

 

      schizophrenic, I worried about today, when he

 

      thanked himself.  But we certainly appreciate all

 

                                                               352

 

      your service and everything.

 

                Thank you.

 

                [Applause.]

 

                MS. WINKLE:  And the other plaque goes to

 

      Marv, and the one thing I want to say about Marv, I

 

      have enjoyed having dinner with Marv in the

 

      evenings.  Last night, for you who weren't at

 

      dinner, he had this huge, huge plate of food, and

 

      he said, "It is actually bigger than it looks."

 

                He has contributed a lot at this meeting,

 

      and we are going to miss him.

 

                [Applause.]

 

                DR. KIBBE:  Marvin, would you like to make

 

      a comment, a last shot across the barrel?

 

                DR. MEYER:  It took I guess 30 years to

 

      get on this committee, but I have thoroughly

 

      enjoyed it.  Everyone around the table brings a

 

      different perspective, and that is what makes it

 

      good for the FDA and fun to be part of.

 

                We have good chairs, Vince Lee, and then

 

      Art Kibbe, and so I would highly recommend this

 

      position for three years to anyone who wishes or

 

                                                               353

 

      gets invited to participate.  Beyond three years

 

      may be questionable.

 

                So, thank you.

 

                DR. KIBBE:  Thank you, Marvin.

 

                I have a whole series of points to make.

 

      First, is that this has been a real joy and an

 

      opportunity to serve and do what I think are useful

 

      things, and to work with people who are dedicated

 

      to having positive outcomes for the American

 

      public.

 

                Most of that, of course, goes, the blame

 

      for how well it turned out goes to Ajaz and Helen,

 

      who lead a great ship and have become close

 

      friends, as well as good working colleagues.

 

                I think we did quite a bit over the years

 

      and I think there is quite a bit more to do, and

 

      the committee needs to move forward, and I would be

 

      happy to help in whatever manner I can.

 

                One of the things that I think you need to

 

      be careful about is that the speed of change is

 

      ever increasing, and like Alice, you are running as

 

      hard as you can just to stay where you are, and to

 

                                                               354

 

      get ahead, you have to jump off of that treadmill

 

      and get on a different path.

 

                One thing that I didn't mention this

 

      morning that you need to keep in the back of your

 

      mind is that according to the U.S. law, treaty

 

      trumps law.  If the Senate and the President want

 

      to sign a treaty with some country that allows for

 

      something to happen, the Food, Drug, and Cosmetic

 

      Act is trumped by the treaty, and whatever rules

 

      and regulations you have, the treaty wins.

 

                Yes, it is absolutely true.  Treaty trumps

 

      law, and the President signs it, and the Senate

 

      agrees to it, and the House of Representatives can

 

      complain all they want, and the regulatory agent

 

      have to readjust.

 

                I would love to see the industry take some

 

      of its money that it spends on direct-to-consumer

 

      advertising and put it into, first, getting the

 

      American public to understand how cost effective

 

      drugs are relative to other therapeutic moieties,

 

      because they don't understand that, because they

 

      see the bill in front of them and they don't see

 

                                                               355

 

      the other bills.

 

                The other thing is to get them to

 

      understand that drugs aren't safe, and they

 

      shouldn't just use them because somebody says it

 

      might be a good deal.  I don't know whether we can

 

      get the industry to do that, because I know that

 

      the ads are meant to sell things rather than not

 

      sell things, and taking out ads to tell people not

 

      to do things is hard to get them to do, but I would

 

      love to do that.

 

                We need to change the criteria for how we

 

      evaluate who well the FDA is doing.  I think there

 

      is way too much pressure on them to produce new

 

      drugs, to produce new reviews, to produce new

 

      things, and I don't know what the right

 

      productivity criteria is.

 

                I know we need to change the productivity

 

      criteria for the U.S. Patent Office, that we have

 

      got to stop them from just issuing patents to make

 

      sure they have issued three patents, and give them

 

      credit for not issuing a patent that shouldn't be

 

      issued.

 

                                                               356

 

                Then, lastly, the goldpost.  When you use

 

      science to establish the goldpost for regulatory

 

      approval, you have moving goalposts because science

 

      moves, science progresses, current best thinking is

 

      always better than it was 10 or 15 years ago, and

 

      both the Agency and the industry have to understand

 

      that that is not a threat, that is an opportunity.

 

                I truly have enjoyed myself and I hope

 

      that what little I have done has contributed to

 

      everybody else having a good time.

 

                I think that it is 4:30 and it is an

 

      appropriate time for us to adjourn.  If there are

 

      not other dramatic statements that need to be made

 

      by anyone else, I will see you next time maybe.

 

                [Whereupon, at 4:30 p.m., the meeting was

 

      adjourned.]

 

                                 - - -