1

 

                DEPARTMENT OF HEALTH AND HUMAN SERVICES

 

                      FOOD AND DRUG ADMINISTRATION

 

                CENTER FOR DRUG EVALUATION AND RESEARCH

 

 

 

 

 

 

 

             ADVISORY COMMITTEE FOR PHARMACEUTICAL SCIENCE

 

 

                               Volume II

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

                         Wednesday, May 4, 2005

 

                               8:30 a.m.

 

 

 

 

 

 

 

 

                CDER Advisory Committee Conference Room

                           5630 Fishers Lane

                          Rockville, Maryland

                                                                 2

 

                              PARTICIPANTS

 

      Charles Cooney, Ph.D., Chair

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

 

      MEMBERS

 

      Patrick P. DeLuca, Ph.D.

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

      Michael S. Korczynski, Ph.D.

      Gerald P. Migliaccio (Industry Representative)

      Kenneth R. Morris, Ph.D.

      Marc Swadener, Ed.D. (Consumer Representative)

      Cynthia R.D. Selassie, Ph.D.

      Nozer Singpurwalla, Ph.D.

 

      SPECIAL GOVERNMENT EMPLOYEES

 

      Carol Gloff, Ph.D.

      Arthur Kibbe, Ph.D.

      Thomas P. Layloff, Jr., Ph.D.

      Marvin C. Meyer, Ph.D.

 

      FDA

 

      Gary Buehler, R.Ph.

      Kathleen A. Clouse, Ph.D.

      Jerry Collins, Ph.D.

      Ajaz Hussain, Ph.D.

      Robert Lionberger, Ph.D.

      Robert O'Neill, Ph.D.

      Keith O. Webber, Ph.D.

      Helen Winkle

      Lawrence Yu, Ph.D.

                                                                 3

 

                            C O N T E N T S

 

                                                              PAGE

      Call to Order

                Charles Cooney, Ph.D.                            5

 

      Conflict of Interest Statement

                Hilda Scharen, M.S.                              5

 

      Parametric Tolerance Interval Test for Dose

        Content Uniformity

 

         Current Update on the Working Group

                Robert O'Neill, Ph.D.                            8

 

      Quality-by-Design and Pharmaceutical Equivalence

 

         Topic Introduction

                Ajaz Hussain, Ph.D.                             10

 

         Using Product Development Information to

           Extend Biopharmaceutics Classification

           System-based Biowaivers

                Ajaz Hussain, Ph.D.                             30

 

         Using Product Development Information to Address

           the Challenge of Highly Variable Drugs

                Lawrence Yu, Ph.D.                              79

 

         Using Product Development Information to Support

           Establishing Therapeutic Equivalence of

           Topical Products

                Robert Lionberger, Ph.D.                       140

 

         Summary of Plan

                Ajaz Hussain, Ph.D.                            169

 

      Committee Discussion and Recommendations                 192

 

      Criteria for Establishing a Working Group for

        Review and Assessment of OPS Research Programs

 

         CBER Peer Review Process for

           Researchers/Reviewers

                Kathleen A. Clouse, Ph.D.                      221

 

         CDER Peer Review Research

                Jerry Collins, Ph.D.                           243

                                                                 4

 

                      C O N T E N T S (Continued)

                                                              PAGE

 

      Committee Discussion and Recommendations                 258

 

      Conclusion and Summary Remarks

                Ajaz Hussain, Ph.D.                            273

                Helen Winkle                                   280

 

                                                                 5

 

                         P R O C E E D I N G S

 

                             Call to Order

 

                DR. COONEY:  I would like to call the

 

      meeting to order this morning.

 

                     Conflict of Interest Statement

 

                MS. SCHAREN:  Good morning.  I am going to

 

      be going through the Conflict of Interest

 

      Statement.

 

                The Food and Drug Administration has

 

      prepared general matters waivers for the following

 

      Special Government Employees:  Drs. Charles Cooney,

 

      Patrick DeLuca, Carol Gloff, Arthur Kibbe, Michael

 

      Korczynski, Thomas Layloff, Marvin Meyer, Kenneth

 

      Morris, Nozer Singpurwalla, who are attending

 

      today's meeting of the Pharmaceutical Science

 

      Advisory Committee, to:

 

                1.  Receive an update on current

 

      activities of the Parametric Tolerance Interval

 

      Level PTIT Work Group;

 

                2.  Discuss and provide comments on the

 

      general topic of considerations for assessment of

 

      pharmaceutical equivalence and product design;

 

                3.  Discuss criteria for establishing a

 

      working group for review and assessment of Office

 

      of Pharmaceutical Science Research Programs.

 

                                                                 6

 

                The meeting is being held by the Center

 

      for Drug Evaluation and Research.  Unlike issues

 

      before a committee in which a particular product is

 

      discussed, issues of broader applicability, such as

 

      the topic of today's meeting, involve many

 

      industrial sponsors and academic institutions.

 

                The committee members have been screened

 

      for their financial interests as they may apply to

 

      the general topic at hand.  Because general topics

 

      impact so many institutions, it is not practical to

 

      recite all potential conflicts of interest as they

 

      apply to each member.

 

                FDA acknowledges that there may be

 

      potential conflicts of interest, but because of the

 

      general nature of the discussions before the

 

      committee, these potential conflicts are mitigated.

 

                With respect to FDA's invited industry

 

      representative, we would like to disclose that Dr.

 

      Paul Fackler and Dr. Gerald Migliaccio are

 

                                                                 7

 

      participating in this meeting as non-voting

 

      industry representatives acting on behalf of

 

      regulated industry.  Dr. Fackler's and Dr.

 

      Migliaccio's role on this committee is to represent

 

      industry interests in general, and not any other

 

      one particular company.  Dr. Fackler is employed by

 

      Teva Pharmaceuticals, Dr. Migliaccio is employed by

 

      Pfizer.

 

                In the event that the discussions involve

 

      any other products or firms not already on the

 

      agenda for which FDA participants have a financial

 

      interest, the 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. COONEY:  Thank you, Hilda.

 

                I am Charles Cooney, the chairman of the

 

      committee, and will preside over the schedule

 

                                                                 8

 

      today.  We have several topics on the schedule.

 

      The first of these is a current update of the

 

      working group Parametric Tolerance Interval Test

 

      for Dose Content Uniformity by Robert O'Neill, who

 

      I believe has just come in.

 

                 Parametric Tolerance Interval Test for

 

                        Dose Content Uniformity

 

                  Current Update on the Working Group

 

                DR. O'NEILL:  I am here to just update you

 

      on what we promised you from the last meeting on

 

      October 19th.  As you know, there is a technical

 

      working group that has been put together with folks

 

      from FDA and folks from the IPAC group.

 

                We have been working diligently since

 

      then.  We thought we would have some

 

      recommendations for you today.  We do not, but we

 

      have had approximately eight get-togethers, five

 

      telecons and three face-to-face meetings, the last

 

      of which was about a week, week and a half ago.

 

                What those discussions have been about is

 

      the agreement of the statistical formulations of

 

      the problem. There were a number of discussions

 

                                                                 9

 

      regarding the coverage probability and symmetry of

 

      coverage, off-target operating characteristic curve

 

      agreements, and things of that nature.

 

                There has been some computer programs that

 

      have been shared back and forth, validation of each

 

      other's methods, and I believe there is now

 

      agreement on that aspect of it and that the working

 

      group is turning towards the agreement on where the

 

      operating characteristic curve ought to be placed

 

      and how it might handle certain particular

 

      situations, particularly off-target means.

 

                That is essentially where we are.  I think

 

      everyone is optimistic that probably the next time

 

      we report to you, that there will be actual

 

      recommendations for you to respond to, but that is

 

      essentially where we are right now.

 

                I would be willing to take any questions

 

      if you have any.

 

                DR. COONEY:  Any comments or questions

 

      from the Committee?  If not, thank you very much

 

      and we look forward to the next step.

 

                DR. O'NEILL:  Thank you.

 

                DR. COONEY:  We will immediately begin

 

      with the second topic this morning, which is

 

      Quality-by-Design and Pharmaceutical Equivalence,

 

                                                                10

 

      to be introduced by Ajaz Hussain.

 

            Quality-by-Design and Pharmaceutical Equivalence

 

                           Topic Introduction

 

                DR. HUSSAIN:  Good morning.  I would like

 

      to introduce to you the Topic No. 2,

 

      Quality-by-Design and Pharmaceutical Equivalence.

 

      We believe we have an opportunity here to explore

 

      and what we plan to do with you is to share some of

 

      our initial thoughts and hopefully, engage the

 

      Advisory Committee in discussion to help us make

 

      sure we are on the right track.

 

                In many ways, the discussions continue

 

      from yesterday where, in essence, we are looking at

 

      opportunities that have been created and

 

      re-examining some of our current policies and to

 

      see how we can realize opportunities to move

 

      towards a desired state.

 

                I would like to put this in the context of

 

      moving from a reactive to a proactive decision

 

                                                                11

 

      system for pharmaceutical quality, and recognizing

 

      that this is only a journey, not a destination.  I

 

      think in the world of continuous learning and

 

      continuous improvement, really, continuous learning

 

      is your destination in some ways.

 

                I think over the last four years, we have

 

      focused on discussing some of the opportunities in

 

      general, but if I look at some of the reactive

 

      examples on this chart, yesterday, in some way, we

 

      talked about testing to document quality, repeating

 

      deviations in our specification investigations, and

 

      in some ways we will start focusing on the other

 

      aspects of prior approval supplement for process

 

      optimization and continuous improvement, multiple

 

      CMC review cycles, but more importantly, I think,

 

      how can we leverage the opportunity of quality by

 

      design for demonstrating therapeutic equivalence of

 

      generic products.

 

                I think we struggle often in this arena,

 

      and we have struggled and unable to sort of find

 

      ways of approving generic products and the degree

 

      of complexity has increased such as topical

 

                                                                12

 

      products and inhalation products, and so forth, so

 

      how can we leverage the pharmaceutical development

 

      information to seek out ways to find mechanisms for

 

      approving complex generic products, I think is a

 

      topic.

 

                I think the proactive examples I think is

 

      quality by design and real time release, right

 

      first time, process optimization and continuous

 

      improvement within the facilities quality system.

 

                But I think in some ways, today, we will

 

      focus on single CMC review cycle, less so, but I

 

      think quality-by- design approach for demonstrating

 

      therapeutic equivalence of generic products would

 

      be a focus.

 

                Therapeutic equivalence.  The definition

 

      from the Orange Book is as follows.  Drug products

 

      are considered to be therapeutic equivalents only

 

      if they are pharmaceutical equivalents and if they

 

      can be expected to have the same clinical effect

 

      and safety profile when administered to patients

 

      under conditions specified in the labeling.

 

                Pharmaceutical equivalent products are

 

                                                                13

 

      drug products are considered to be pharmaceutical

 

      equivalents if they contain the same active

 

      ingredients, are of the same dosage form, route of

 

      administration, and are identical in strength or

 

      concentration.

 

                Pharmaceutically equivalent drug products

 

      are formulated to contain the same amount of active

 

      ingredient in the same dosage form  and to meet the

 

      same or compendial or other applicable standards of

 

      strength, quality, purity, and identity, but they

 

      may differ in characteristics such as shape,

 

      scoring configuration, release mechanisms,

 

      packaging, excipients, expiration time, and within

 

      certain limits, labeling.

 

                So, I think we have certain flexibility

 

      built into the issue of pharmaceutical equivalence,

 

      and one of the desired states is to leverage that

 

      and to say that we want to set specification based

 

      on mechanistic understanding, so if you have a

 

      different mechanism, that is perfectly fine, but

 

      then you set your specification on that.

 

                The discussion yesterday was not to force

 

                                                                14

 

      the generic to a particular specification, but

 

      recognize that as part of that.

 

                Today, I think I would like to put this in

 

      the context of risk, uncertainty, and variability,

 

      and I think that framework will help us think more

 

      clearly about the issues.

 

                FDA classifies as therapeutically

 

      equivalent those products:  That are approved safe

 

      and effective, so you have to have a reference

 

      product which is safe and effective, and has an

 

      approved pharmaceutical equivalents against the

 

      repetition of that, and are bioequivalent, that

 

      they do not present a known or potential

 

      bioequivalence problem, and they meet an acceptable

 

      in vitro standard, or if they do present such a

 

      known or potential problem, they are shown to meet

 

      an appropriate bioequivalence standard.

 

                In absence of pharmaceutical development

 

      information and quality-by-design aspect, we have

 

      to assume that they present a bioproblem, so we

 

      often go to the second bullet in most cases; are

 

      adequately labeled, and are manufactured in

 

                                                                15

 

      compliance to Current Good Manufacturing Practice

 

      regulations.

 

                In a sense, the last four years, our

 

      initiative goals have been to focus on

 

      science-based, risk-based approaches, and there are

 

      certain challenges I think in our articulation of

 

      the problem today.

 

                I want to sort of share with you what the

 

      challenges might be.

 

                Risk-based scientific decisions on

 

      pharmaceutical quality is clearly our goal.  Risk

 

      is a combination of the probability of occurrence

 

      of harm and the severity of that harm.  The reason

 

      for focusing on that is an aspect that I feel that

 

      we often get entangled in and unable to really

 

      articulate the problem carefully.

 

                Uncertainty with respect to severity of

 

      harm and/or probability of its occurrence and their

 

      modulating factors is that challenge that we face,

 

      what are the critical quality attributes, and what

 

      is an acceptable variability.

 

                I have argued, and I think I will ask the

 

                                                                16

 

      Committee to think about this, is my argument sort

 

      of correct, in some ways, in the current decision

 

      system which tends to be reactive, one contributing

 

      factor for the reactive decision system is we

 

      confound uncertainty, variability, and risk.

 

                This is, by nature, how we develop our

 

      products to a large degree.  The current paradigm

 

      for product and process development tends to do

 

      this, because our entire effort in new drug

 

      development, for example, is focus on safety and

 

      efficacy of a molecule, but we use a product to

 

      achieve that.

 

                The connection between product quality and

 

      the clinical is generally focused on the molecules

 

      rather than the product.  So, that is a part of

 

      this discussion.

 

                Often intrinsic safety and efficacy of a

 

      new molecular entity is confounded with its product

 

      and manufacturing process just by the nature of our

 

      product development and process itself.

 

                We have multifactorial aspects of

 

      pharmaceutical products and manufacturing

 

                                                                17

 

      processes, and there is increasing complexity, and

 

      if we can find a way to articulate our problem more

 

      carefully, this may help moving forward more

 

      quickly.

 

                Establishing constraints based on prior

 

      knowledge and limited development experiments that

 

      are done in the development cycle.

 

                What I have argued is there is a need to

 

      entangle or, as I call it, de-convolute

 

      uncertainty, variability, and risk, and then to

 

      achieve truly a scientific integration of these for

 

      quality decisions, how we set specification.

 

                Yesterday, for example, what we proposed

 

      was an assessment of variability, example, begins R

 

      and R.  That is an attempt to start characterizing

 

      the variability and attempt to start teasing out

 

      what comes from what.

 

                This may appear to be paradoxical, and it

 

      probably is without the concept of quality by

 

      design, and that sort of links to Dr. Woodcock's

 

      paper that we talked about.

 

                Let me illustrate that.  When we approve a

 

                                                                18

 

      new drug application, we bring assessment of all

 

      the disciplines to bear on a decision which says

 

      the risk to benefit ratio of this proposed drug

 

      product is acceptable when used in accordance to

 

      the label.  That is the pivotal decision criteria.

 

                Now, that is based on the pivotal clinical

 

      trials along with the toxicology, and along with

 

      all the studies that go along with that, but the

 

      pivotal clinical trials play the major role there.

 

                What do we use for the pivotal clinical

 

      trials?  We use a product, and we often do not have

 

      the opportunity or do not have the intent to gauge

 

      whether the product is modulating the safety and

 

      efficacy of the molecule beyond that of exposure

 

      less the bioavailability.

 

                So, from that aspect, the quality of the

 

      product has to be built in before you get into the

 

      clinical trials, otherwise you confound the

 

      clinical trials with quality problems, and

 

      actually, I will illustrate one example in my

 

      second presentation of that.

 

                As that happens, then, the product is

 

                                                                19

 

      approved and then manufacturing is transferred to

 

      production and you have seven production lots, but

 

      that goes to the patient population.

 

                So, you see the disconnect between

 

      pharmaceutical quality or product quality.  I am

 

      not talking about the safety and efficacy of a

 

      molecule, I am talking about how a product

 

      modulates that.

 

                So, in many ways, if I look at drug

 

      development program, and here is an illustration of

 

      that, this is actually a real case study.  The only

 

      difference I have here is I have a line that made

 

      it more linear.  That is the only change I did, but

 

      here is a development program.

 

                The initial formulation was a capsule.

 

      Then, they went to a table, wet granulation tablet,

 

      and each star is a bioequivalent study.  So, they

 

      qualified that change from capsule to tablet using

 

      a bioequivalent study.  Then, they add a film coat,

 

      then, change the site of manufacture of the drug

 

      substance, and so forth.

 

                You see the changes that were occurring in

 

                                                                20

 

      this drug development program, some before clinical

 

      trials were initiated, some during the clinical

 

      trials, site changes, and so forth.

 

                But this company actually qualified, what

 

      they call bridging studies using a bioequivalence

 

      trial.  Towards the end of the review time, to

 

      qualify and bridge the clinical trial, pivotal

 

      clinical trial material with the to-be-marketed

 

      product, they opted to use multiple-dose studies of

 

      the traditional thing, and that failed at the last

 

      minute.  Actually, it didn't fail, it just failed

 

      to establish bioequivalence, so they went back and

 

      actually repeated the study with a larger number of

 

      subjects, but it delayed them.

 

                The point here is all these changes are

 

      being qualified on the basis of their traditional

 

      bioequivalence trial.  In new NDAs, we generally

 

      see from 3 to 6 clinical bioequivalence studies,

 

      and that was our number.

 

                Here is a number that was shared with us

 

      by Jack Cook sometime, this is the year 2000, about

 

      7 per approved compound.  In a sense, if I look

 

                                                                21

 

      at--this is Gary Buehler's slide--I think in many

 

      ways, the difference between the generic drug

 

      approval and the new drug approval is minimal when

 

      it comes to bioequivalence trial.

 

                In many ways, we use bioequivalence to

 

      gauge information which we don't have in the NDA,

 

      but in reality, in some cases, it is simply the two

 

      are comparable.  So, from that aspect, I think when

 

      I look at oral products, immediate release

 

      products, and here is my demarcation in an attempt

 

      to sort of categorize what is uncertainty, what is

 

      variability, and what is risk.

 

                The goal of our generic drug approval

 

      process is to approve a generic product.  An

 

      approved product is expected to have the same

 

      clinical effect and safety profile when

 

      administered to patients under conditions specified

 

      in the labeling.

 

                Based on the previous slide, I have shown

 

      the characteristics of this decision system is,

 

      one, the first one, the product must be

 

      pharmaceutically equivalent, and here the questions

 

                                                                22

 

      are has the applicant demonstrated that it's the

 

      same active, identical amount, same dosage form,

 

      route of administration, and so forth, identity,

 

      strength, quality, purity.

 

                So, that is an assessment process of how

 

      good the identical methods are and how good they

 

      have qualified, so it's a knowledge-based decision,

 

      there is an uncertainty aspect associated with

 

      that.

 

                Then, we have to define acceptable

 

      variability for that product, and we rely on the

 

      compendial or other standards to do that.

 

                The risk in this case, I am talking about

 

      risk from a clinical perspective, is the prior

 

      knowledge that come from NDA.

 

                Need for bioequivalence assessment for

 

      oral products is that next question, and again the

 

      same words from the Orange Book is if you do not

 

      present a known or potential bioequivalence

 

      problem, acceptable in vitro standard is fine.  But

 

      you saw the debate with dissolution, we often don't

 

      go there, we often go to a bioequivalence study.

 

                If you go to in vitro standard compendial

 

      dissolution test method, if you go to a

 

      bioequivalence standard, then, the acceptable

 

                                                                23

 

      variability is our bioequivalence standard, 90

 

      percent confidence interval for test or reference

 

      ratio for rate and extent of absorption is within

 

      80 to 125 percent.

 

                It has to be adequately labeled and

 

      manufactured in conformance to cGMPs, and in that

 

      case, acceptable variability is what we tolerate in

 

      terms of deviation or specifications, and so forth.

 

                So, that is one way of looking at trying

 

      to partition uncertainty, variability, and risk, so

 

      that we can formulate the right questions.

 

                Clearly, I think the quality-by-design

 

      thinking is intended to focus on the intended use,

 

      and design is about doing things consciously.  I

 

      showed this slide to you before.

 

                I think what we would like to consider is

 

      in the context of pre- and post-approval changes,

 

      generic drugs, and even extending that to the

 

      concept of follow-on protein pharmaceuticals, the

 

                                                                24

 

      primary goal of a scientific decision framework--I

 

      am not talking with a regulatory process--the

 

      decision criteria that we bring, need to understand

 

      the complexity and uncertainty, but the decision

 

      process should be consistent.

 

                I think that is the fundamental basis that

 

      we work under.  Furthermore, I think our goal is

 

      also to identify and eliminate unnecessary human

 

      and animal testing for this decision framework,

 

      keeping in mind that most bioequivalent studies are

 

      done in normal healthy subject volunteers, new

 

      drugs and so forth.

 

                If we can avoid exposing normal healthy

 

      subject human volunteers, it is desirable, and that

 

      is part of the regulation.  I will share that

 

      regulation with you.

 

                So, Topic 2 today, the premise that we had

 

      in mind was that a quality-by-design approach via

 

      pharmaceutical development information can

 

      potentially provide an excellent means to address a

 

      number of challenges previously discussed at ACPS

 

      meetings without complete or satisfactory

 

                                                                25

 

      resolution, for example, bioequivalence of highly

 

      variable drugs, bio-in-equivalence criteria,

 

      pharmaceutical and therapeutic equipment of locally

 

      acting drug products, such as topical products.

 

                Today, Lawrence will bring his thoughts to

 

      you, and these are our preliminary thoughts, and I

 

      think we just wanted to put our preliminary

 

      thoughts on the table to engage you and engage the

 

      community to help us think about this, so that as

 

      we spend our time thinking about this, we already

 

      have some feedback and we are also on the right

 

      track.

 

                Rob Lionberger will come back.  He

 

      presented a decision tree to you before, but he

 

      will recast that decision tree for topical products

 

      in the context of quality-by-design.

 

                Yesterday, we focused on dissolution

 

      testing, and as the past chair of the BCS Working

 

      Group, I took it upon myself to sort of go back and

 

      re-examine what were my personal thoughts and what

 

      held me back to make the recommendations that you

 

      see in the guidance and see how we can recast that

 

                                                                26

 

      discussion into the quality-by-design thinking, so

 

      that I can take this discussion and give that as a

 

      recommendation to the current Technical Committee

 

      on Biopharmaceutical Classification System.

 

                Again, as I said, these are initial

 

      thoughts, and our goal is to engage you in a debate

 

      and discussion to hopefully give us some

 

      perspective are we on the right track.  The three

 

      topics today are:

 

                How can pharmaceutical development

 

      information help to extend the applications of

 

      BCS-based waivers of in-vivo studies for immediate

 

      release products?

 

                How can pharmaceutical development

 

      information be utilized to address the challenges

 

      of highly variable drugs?

 

                How do we use this to establish

 

      therapeutic equivalence of topical products?

 

                Those are the three topics that we would

 

      like to present, and the general question is are we

 

      on the right track, but then more detailed

 

      recommendations on how we should proceed with these

 

                                                                27

 

      three topics or other topics that we should have

 

      considered instead of this.

 

                So, that is the Topic 2.

 

                DR. COONEY:  Thank you, Ajaz.

 

                We might pause for a moment for any

 

      questions particularly around clarification of

 

      these opening comments from the committee.  Art?

 

                DR. KIBBE:  The question I always struggle

 

      with is how do we define highly variable drugs.

 

      Are we defining them in clinical outcomes, because

 

      then the dosage form might not be involved in it at

 

      all.

 

                DR. HUSSAIN:  The definition hinges on the

 

      bioequivalence drug, the variability that we

 

      estimate from the bioequivalence drug.

 

                DR. KIBBE:  But that could be a function

 

      of intersubject variability, subject population

 

      selection, and have nothing at all to do or

 

      minimally to do with the actual product that you

 

      are making.

 

                DR. HUSSAIN:  That is the discussion

 

      Lawrence will bring to you, so if you hold that

 

                                                                28

 

      question for Lawrence.

 

                DR. KIBBE:  Thank you.

 

                DR. COONEY:  Ken?

 

                DR. MORRIS:  The two comments is that the

 

      Topic 2 premise, the other part of that premise is

 

      that the proper development information is being

 

      generated at the companies which, of course, you

 

      have limited control over, and that is being

 

      shared, just as a caveat.

 

                DR. COONEY:  Nozer?

 

                DR. SINGPURWALLA:  Two comments.  Slide

 

      No. 7.

 

                DR. HUSSAIN:  I am sorry, I don't have

 

      numbers.

 

                DR. SINGPURWALLA:  I know.  Components of

 

      the Challenge.  The second bullet.  That second

 

      bullet is wrongly worded, it has to be changed, and

 

      I will tell you why.

 

                The more important reason is you, on your

 

      eight bullet, are talking about confounding of

 

      uncertainty, variability, and risk.  They should

 

      not be confounded.  Who is confounding them and

 

                                                                29

 

      why?

 

                DR. HUSSAIN:  The current system has a

 

      tendency to do that.  That is what I mean.

 

                DR. SINGPURWALLA:  But that simply means

 

      that the system is not educated enough, because

 

      variability is the cause of uncertainty, is one of

 

      the causes of uncertainty, and risk is a measure,

 

      is a way to measure uncertainty and its

 

      consequence.

 

                So, why is there so much confusion about

 

      these very elementary ideas in the industry and

 

      perhaps in the pharmaceutical community?

 

                DR. HUSSAIN:  I don't know how to answer

 

      that.

 

                DR. SINGPURWALLA:  Well, they need to be

 

      trained, educated.

 

                DR. HUSSAIN:  But let me propose this in

 

      the sense, uncertainty is not risk.

 

                DR. SINGPURWALLA:  I agree with you.  I

 

      agree with what you are saying completely.  What I

 

      am asking is, what is the cause of this confusion,

 

      and it is so easy to remove this confusion?

 

                DR. HUSSAIN:  I understand the concern you

 

      are expressing, and my premise is for years we have

 

      not utilized the pharmaceutical development, and we

 

                                                                30

 

      have treated that as an art rather than a science,

 

      and that is the way to get away from that

 

      confusion.  So, that is the premise.

 

                DR. COONEY:  Any other comments at this

 

      point?  We will have ample opportunity for further

 

      discussion.

 

                Using Product Development Information to

 

                Extend Biopharmaceutics Classification

 

                        System-Based Biowaivers

 

                DR. HUSSAIN:  Let me go on to the

 

      Biopharmaceutics Classification System.  In

 

      preparing for this presentation, I actually went

 

      back and reviewed all the publications that have

 

      occurred in this arena in this discipline, in this

 

      topic area for the last five years, and there has

 

      been a tremendous number of progress in this area.

 

                For example, more recently, Professor Les

 

      Benet's article was published on how you can

 

      actually start predicting metabolism, and so forth,

 

                                                                31

 

      and how you can sort of add that additional

 

      dimension.

 

                There has been a paper published in Pharm

 

      Research on quantitative instead of, you know,

 

      rigid boundaries, and so forth.

 

                But instead of sort of trying to summarize

 

      the progression signs, what I wanted to do was to

 

      go back and re-examine my own thoughts that were

 

      expressed to the Advisory Committee in the year

 

      2000, so I am actually going to repeat an old

 

      presentation, but in light of what we have learned

 

      in the last four years.

 

                My goal here is to share with you some of

 

      the concerns we had when we proposed the BCS-based

 

      waiver guidance in the year 2000, and to what

 

      extent those concerns remain, and to what extent

 

      quality-by-design may be able to alleviate some of

 

      this concern, and the discussion with you, I intend

 

      to use that as recommendation to the current

 

      Technical Committee on BCS.  So, that is the game

 

      plan.

 

                This is an old presentation with some

 

                                                                32

 

      minor modification.  When I had made this

 

      presentation, I was completely focused on risk, and

 

      the title was "Biopharmaceutics Classification

 

      System: A Risk Management Tool."

 

                In light of the learning that I at least

 

      personally had, I want to sort of bring in the

 

      uncertainty and variability components to this.

 

                Since this is a presentation, probably my

 

      last presentation on the BCS topic before I handed

 

      over the reins of responsibility to Lawrence and

 

      Mehul, the new BCS Technical Committee was formed,

 

      when Helen asked me to move to OPS and the PAT

 

      process got started, so my focus went to PAT for a

 

      reason which connects back to this one.

 

                So, this BCS Technical Committee has been

 

      in place under the leadership of Lawrence and

 

      Mehul, and they have been diligently addressing a

 

      number of implementation issues trying to

 

      coordinate all the submissions, and so forth, and

 

      there has been significant activity on this

 

      guidance on the new drug side, very little, if any,

 

      on the generic side.

 

                You also heard from Mehul the database is

 

      now almost ready, is being audited, database and

 

      prospective research for extensions, links to PQRI

 

                                                                33

 

      and FIP, but the PQRI program really didn't take

 

      off, and our thoughts were we wanted to explore

 

      extension of BCS-based biowaivers to Class III and

 

      Class II drugs.

 

                Further research at the FDA, which we

 

      completed, and we did extend the BCS-based

 

      biowaivers to "fed" bioequivalence studies, and

 

      that was part of the thing, and that work was done

 

      with collaboration with Tennessee.

 

                Continuing education initiatives and

 

      practitioners and public, and the group has been

 

      busy.  International harmonization was an aspect,

 

      but to the extent the definition of high solubility

 

      and rapid dissolution, we got into ICH Q6A, the

 

      European Guideline also adopted much of the BCS

 

      recommendation to some extent.  There are certain

 

      differences, though, it is not fully harmonized.

 

                With that as a background, let me trace

 

      for you the evolution of the recommendations in the

 

                                                                34

 

      BCS guidance that we released in the year 2000.

 

                Regulatory Bioequivalence: An Overview,

 

      from my perspective, this is a graphical

 

      representation of our regulation.

 

                If you look at the dosage form that we

 

      deal with, solutions, suspensions, chewable

 

      tablets, conventional tablets, and modified release

 

      products, for solutions, we consider bioequivalence

 

      essentially is self-evident, bioavailability is

 

      self-evident, and biowaivers are granted, condition

 

      being excipients do not alter absorption, and that

 

      is an assessment based on historical data.

 

                For any product that contains drug in a

 

      solid form, we have a concern, and for pre-1962

 

      drugs, we call DESI drugs, in vivo evaluation for

 

      bio-problem, that was the original biopharm

 

      classification system, if you really look at it,

 

      that had many of the elements of therapeutic index,

 

      PK, the solubility, and so forth.

 

                For post-1962 drugs, generally, in vivo,

 

      some exceptions with IVIVC.  Then, we introduced a

 

      SUPAC-IR guideline in '95, and we introduced the

 

                                                                35

 

      elements of BCS guidance, BCS system in that to

 

      give a waiver for minor changes and moderate

 

      changes that Mehul talked to you about yesterday.

 

                For modified release, we don't have a

 

      classification system, bioequivalence has to be

 

      demonstrated in vivo with the exception of SUPAC

 

      modified release for within a product.  If you have

 

      in vitro bioequivalence, you can make changes.

 

      Again, Mehul summarized that.

 

                I want to trace back the discussion to a

 

      bioequivalence hearing, which I did not attend.  I

 

      was just graduating in '86, but I was connected to

 

      this because I made the slides of a number of

 

      people who presented here, so I knew what was

 

      happening.

 

                This was a pivotal discussion and I think

 

      set the stage for what evolved as bioequivalence

 

      standard.  There were two comments that I want to

 

      share with you.

 

                One was Dr. Bob Temple.  He said, after

 

      the end of this discussion, it seems sensible to

 

      think that swallowing something that turns into a

 

                                                                36

 

      solution rapidly would be difficult to lead to

 

      differences from one product to another.

 

                So, the clinicians were arguing you don't

 

      need biostudies for everything.  Arnold Beckett had

 

      made that argument years at that time, so he said

 

      you shouldn't go with in vivo for everything, but

 

      we did.

 

                Milo Gibaldi, an eminent pharmacokinetic

 

      professor, "I have learned that there is no support

 

      here for attempting to provide such assurance

 

      solely with in vitro data."  So, that was a pivotal

 

      aspect, I think, and I went back and sort of tried

 

      to examine the thoughts and the concerns that were

 

      expressed at that session.

 

                The other aspect that I do want to put on

 

      the table is need to reduce our reliance on in vivo

 

      bioequivalence studies.  Why?  Ethical reasons.  21

 

      CFR 320.25 says, "No unnecessary human research

 

      should be done." Science continues to provide new

 

      methods to identify and eliminate unnecessary in

 

      vivo bioequivalence studies.

 

                Focus on prevention, "building quality

 

                                                                37

 

      into products - right first time."  So you see the

 

      PAT initiative and how this will connect to that

 

      was in the thought process and why we aggressively

 

      moved in that direction, "right first time," I used

 

      before Pfizer.

 

                Time and cost of drug development and

 

      review is a key, because if you see that we have

 

      three to six bioequivalent studies in our NDAs.  We

 

      actually at some point said we don't even review

 

      some of those because they are redundant, so why

 

      expose normal healthy subjects to a new drug which

 

      is under development with all the risks associated

 

      with that.

 

                So, prior to SUPAC-IR/BCS, in vivo

 

      bioequivalence assessments to justify a majority of

 

      manufacturing changes.  So, this was a significant

 

      hurdle, and that changed.  In the SUPAC-IR guidance

 

      in '95, we brought in the classification system and

 

      provided a tiered approach for changes based on in

 

      vitro.

 

                For example, highly soluble, highly

 

      permeable drug, the critical processes for gastric

 

                                                                38

 

      emptying, dissolution is not likely, and

 

      dissolution is not likely to be rate limiting, but

 

      we said 0.1 single point, 85 percent, and so forth.

 

                So, you can see that for each class, we

 

      recommended a tiered approach for waiver of

 

      biostudies for minor changes, and so forth.  We

 

      excluded the Narrow Therapeutic Index drugs from

 

      waiver consideration, but we never defined what

 

      narrow therapeutic index was, and we still haven't.

 

                The guidance in 2000 really extended that

 

      and put that as a waiver for first time approval,

 

      and also provided the methods to classify your

 

      drug, and so forth.  The pivotal recommendation in

 

      that was waiver for in vivo bioequivalence studies.

 

                I do want to stop here and say the title

 

      of this guidance was debated to the nth degree

 

      before we agreed on this internally.  The word

 

      "waiver" was to signify that we want an in vivo

 

      study for everything that is in solid, so the title

 

      was very carefully chosen to reflect that.

 

                Anyway, it's waiver for in vivo

 

      bioequivalence studies is recommended for a solid

 

                                                                39

 

      oral test product that exhibit rapid and similar in

 

      vitro dissolution under specified conditions to an

 

      approved reference product when the following

 

      conditions are satisfied:  products are

 

      pharmaceutically equivalent, drug substance is

 

      highly soluble and highly permeable and is not

 

      considered to have a narrow therapeutic range and

 

      excipients used are not likely to affect drug

 

      absorption.

 

                The class membership, the boundaries that

 

      you see, which are rigid, high solubility, the

 

      highest dose strength is soluble in less than or

 

      equal to 250 ml of aqueous buffers over the pH

 

      range that we had 1 to 6.5 or so, whatever that

 

      thing is I forgot.

 

                The reason for 250 ml is the glass of

 

      water that we take when we do a bioequivalence

 

      study.  High permeability, the extent of absorption

 

      in humans is determined to be greater than 90

 

      percent.

 

                Rapid dissolution is 85 percent dissolves

 

      within 30 minutes in three different conditions,

 

                                                                40

 

      HCL, pH 4.5 and 6.8 buffers using traditional

 

      settings of dissolution apparatus.

 

                Now, clearly, I had approached this as a

 

      risk to bio-in-equivalence because since we started

 

      with the premise that you needed bioequivalence

 

      trials for approval of changes, and so forth, so

 

      the risk factor for me was the proposal the

 

      recommendations should not result in

 

      bio-in-equivalence.

 

                The risk factors that we had in mind were

 

      clearly manufacturing changes, poor process

 

      capability, high between and within batch

 

      variability, but the thing we focused on, reliance

 

      on in vitro dissolution tests and how reliable that

 

      is, single point specification, sampling,

 

      predictability were the issues.

 

                Clearly, the other factors were there,

 

      deficiencies in BE study design, Type II errors,

 

      and so forth.

 

                Now, assessment of risk, what is the risk

 

      of bio-in-equivalence between two pharmaceutically

 

      equivalent products when in vitro dissolution test

 

                                                                41

 

      comparisons are used for regulatory decisions?

 

      That was the question we asked.

 

                What is the likelihood of occurrence and

 

      the severity of the consequences?

 

                Severity was not meeting the

 

      bioequivalence criteria was unacceptable, but what

 

      was the likelihood, so we needed some information

 

      on that.

 

                Regulatory decision, whether or not the

 

      risks are such that the project can be pursued with

 

      or without additional arrangements to mitigate the

 

      risk, and clearly, acceptability of the decision,

 

      is the decision acceptable to society?  This took

 

      four years.

 

                We started working on this in '96, and if

 

      you think I was busy with the PAT presentations

 

      around the globe, that is exactly had to do the

 

      same thing for this one, too, because the mind-set,

 

      the paradigm was so entrenched in the old system.

 

                Professor Gordon Amidon spent some time

 

      with us, he and I.  I had the luxury of having the

 

      biopharm document room right outside my office in

 

                                                                42

 

      the Parklawn Building, so we went through a number

 

      of applications, about 160 applications at that

 

      point, to get a sense of what is happening.

 

                Roughly, what we found was on the new drug

 

      side only, because we have failed studies or we

 

      have all the studies submitted on the generic side,

 

      we couldn't use that database because you just have

 

      the passing studies in there.

 

                So, we looked at the new drug side and

 

      said when does dissolution signal

 

      bio-in-equivalence or does not signal

 

      bio-in-equivalence.  What we found there was

 

      generally, you see big differences in dissolution,

 

      no difference in blood levels.

 

                But, on the other hand, there were signals

 

      that dissolution fails to signal bio-in-equivalence

 

      about 30 percent of the time, and we wanted to ask

 

      why.

 

                So, minimizing risk of bio-in-equivalence,

 

      does in vitro dissolution process emulate in vivo

 

      dissolution process in vitro and in vivo?  Dosage

 

      form disintegration, dissolution, and stability

 

                                                                43

 

      were the concern.

 

                The gastrointestinal fluid volume,

 

      composition, and hydrodynamic conditions were the

 

      concern, and clearly, I think one thing which was

 

      pivotal for the oral discriminating part was the

 

      surface tension, and that could have been picked

 

      up.

 

                Residence time in the stomach and small

 

      intestine were an issue, so we did a lot of

 

      analysis actually of gastric emptying and what

 

      factors affect gastric emptying, and so forth.

 

                Impact of excipient differences on GI

 

      physiology and drug bioavailability were the

 

      questions.

 

                The key question was how well this

 

      emulates in vivo, because this is our standard

 

      dissolution test.

 

                This was a cartoon that I prepared and to

 

      take a look at typical physiologic parameters in a

 

      single dose fasting BE study.  We had fairly good

 

      estimates of the gastric fluid plus the 8 ounces of

 

      water.  We knew what the gastric pH range is

 

                                                                44

 

      generally in the normal subject.

 

                We had the information on the gastric

 

      emptying time, which is highly variable, but

 

      approximately T50 is 15 percent.  The permeability

 

      is low, and that was an advantage in the stomach

 

      compared to small intestine, the surface tension is

 

      lower, and clearly, volumes in the small intestine

 

      were uncertain, and pH, and so forth, and the

 

      permeability was high.  Hydrodynamics was a big

 

      question in our minds.

 

                Lawrence summarized to you the debates

 

      that we have had for dissolution for the last 30

 

      years, and that dissolution tests are over

 

      discriminating, on one hand, and in the USP, the

 

      statement that products that dissolve about 70

 

      percent in 45 minutes have no medically relevant

 

      bioequivalence problems, what was the basis of

 

      that.

 

                Dissolution tests are not sufficient to

 

      assure bioequivalence, and demonstration of IVIVC

 

      is necessary, but when you do that, product

 

      specific, so those are two sides of the debate.

 

                I showed you this slide of the problems

 

      with the dissolution tests of false positives and

 

      false negatives, but then we also looked at things

 

                                                                45

 

      that we made decisions on.

 

                Here is a product.  The generic product

 

      was Product B.  We actually withdrew this product

 

      from the market after approval.  This is a pre-62

 

      drugs, it was approved on the basis of dissolution,

 

      meeting the USP specification criteria.

 

                This was a pre-62 drug, one of the older

 

      drugs, and we had a challenge from the innovator,

 

      which is Product A, that the generic is not

 

      bioequivalent, and that was the basis at which we

 

      had withdrawn this product, Product B, from the

 

      market.

 

                The Cmax, you can see is clearly high, but

 

      in many ways, Product B was more reliable, less

 

      variable, and it was more modern technology, but

 

      the constraints on us is you have to be equal, if

 

      not better.

 

                Here are examples of where there were real

 

      dissolution impacts on in vivo absorption.  Here is

 

                                                                46

 

      a weak acid where the initial formulation for

 

      clinical was capsule.  They went to

 

      wet-granulation, and the to-be-marketed was a

 

      direction compression with dicalcium phosphate, and

 

      the dissolution in this case was Q17 30 minutes in

 

      simulated intestinal fluid.

 

                That's the criss-cross you see if you do

 

      acid in alkaline conditions, you don't distinguish

 

      that, and this had to be reformulated.  But this

 

      was I think in my mind a signal that we probably

 

      are designing our products for dissolution rather

 

      than the intended use.

 

                I wanted to walk through this with you,

 

      and that was one of the reasons for the

 

      quality-by-design thinking.

 

                Here is a Drug X.  This is actually a

 

      clinical study, 100 mg dose, so it's a highly

 

      soluble drug by all definition.  It's a weak base

 

      exhibits a sharp decline in solubility with

 

      increasing pH above 3.

 

                Now, the clinical trial material in this

 

      case was wet granulation, drug particle size of D50

 

                                                                47

 

      of 80 microns, and this was a concern, because our

 

      particle size specification was very crude.  I mean

 

      what does D50 percent give me.  We had lactose,

 

      microcrystalline cellulose, and so forth.  You see

 

      that formulation there.

 

                But the point to focus there is the

 

      dissolution 0.1 normal HCL was 65 percent in 15

 

      minutes and 100 percent in 20 minutes.

 

      Disintegration time was 10 minutes.

 

                The way I had presented this, the

 

      to-be-marketed was the formulation of direct

 

      compression, but actually the wet granulation in

 

      this case was a U.S. formulation, a formulation

 

      using U.S. clinical trials, and the European

 

      clinical trials were done with the to-be-marketed,

 

      and we had to bridge those together.

 

                The to-be-marketed formulation, you can

 

      see what happened here.  Direction compression,

 

      drug particle size of 300 microns, dicalcium

 

      phosphate, and so forth, and the dissolution is

 

      more rapid now, 0.1 normal HCL - 85 percent in 15

 

      minutes, about 95 percent in 20 minutes, so the

 

                                                                48

 

      initial dissolution burst is very rapid,

 

      disintegration time is 1 minute.

 

                Clinical product exhibits poor dissolution

 

      at 7.4. Can you imagine, I mean this is a BCS Class

 

      I drug.  The Cmax or the rate or the exposure of

 

      this in terms of p concentration for the

 

      to-be-marketed formulation in this case was half,

 

      so you see half the blood levels in terms of

 

      height.  So, it simply was signal that if you don't

 

      get the physicochemical properties of drug

 

      understood, you will have these problems.

 

                So, in vitro and in vivo dissolution,

 

      dissolution methods evolved over the last 30 years.

 

      The year 2000, I said there were reproducible test

 

      methods for lot-to-lot quality assurance, so you

 

      can imagine my surprise of the calibration, which I

 

      was not aware of at that time.

 

                The dissolution media volume and

 

      composition selected to maintain "sink" conditions.

 

      In vivo dissolution is a complex process, and you

 

      have to consider pH profile, bile concentration,

 

      motility patterns, and so forth.

 

                In vivo, the "sink" condition is created

 

      due to intestinal permeability, and this was a

 

      contention, which Lawrence and others, we recently

 

                                                                49

 

      published to show how permeability actually impacts

 

      in vivo dissolution, so we have published on this

 

      now already.

 

                I will talk to you about in vitro-in vivo

 

      correlation.  The formulation specificity of IVIVC

 

      has been known since 1997.  This is drug

 

      spironolactone from a publication in J. Pharm

 

      Science in '97.

 

                So, you can see a change in particle size.

 

      You may have a correlation, but a change in

 

      particle size could be outside that.  So, a

 

      correlation itself why this formulation specific

 

      has to be really brought into context.

 

                So, reliance on current dissolution

 

      practice can pose an unacceptable level of risk

 

      from bio-in-equivalence perspective.  Compared to

 

      high solubility drugs, risk is higher for low

 

      solubility drugs.

 

                Products with slow or extended dissolution

 

                                                                50

 

      profiles pose a higher risk.  The dissolution is

 

      rate limiting.

 

                So, in a sense, we wanted to use

 

      dissolution test only to rule out that dissolution

 

      is not rate limiting.  So, that was the basis for

 

      our thought process.  So, we constructed a rapid

 

      dissolution criteria for that purpose.  We did not

 

      want to use dissolution tests for bioavailable

 

      decisions if there was a hint that dissolution is

 

      rate limiting.

 

                Potential for significant differences

 

      between in vivo and in vitro "sink" conditions

 

      higher for low permeability drugs, which we had to

 

      prove later on with a simulation study that

 

      Lawrence did.

 

                Now, to establish a boundary for rapid

 

      dissolution, we simply postulated that since

 

      gastric mucosa does not have high permeability to

 

      drugs, you have a 15-minute time, so you can take

 

      advantage of that.

 

                So, if the dissolution is rapid, then,

 

      much of it is complete before it empties into the

 

                                                                51

 

      small intestine where you have high permeability.

 

      So, in a sense, a very rapidly dissolving drug will

 

      behave essentially like a solution, and it does.

 

                So, here is a snapshot of dissolution

 

      versus AUC and Cmax ratios and the bioequivalence

 

      goalpost of a drug metoprolol.  The reason I did

 

      not take the name off here, because this is already

 

      an ACPS presentation, it is already on the website.

 

                So, you can see a solution versus all the

 

      other formulations that we have approved, generic

 

      and innovator, plus there are research

 

      formulations, which we deliberately made to be very

 

      slowly dissolving.

 

                You see that essentially, for the most

 

      part, the slope is zero.  We then did extensive

 

      simulation work to establish that if in vivo

 

      dissolution is rapid, as a function of different

 

      gastric emptying as a function of mean intestinal

 

      transit time, you are not likely to see a

 

      difference between solution and a tablet, and that

 

      was the basis for our 85 percent and 30 minute in

 

      vitro criteria for rapid dissolution, but we did

 

                                                                52

 

      not apply that to low permeability drugs because of

 

      the risk factors that we felt were coming from

 

      excipients.

 

                The question we asked was is the current

 

      approach for evaluating excipients for decisions

 

      related to biowaiver for oral solutions sufficient.

 

      That is the database I have.

 

                There were hints that the excipient

 

      effects were not fully appreciated.  For example,

 

      there was a study by Ian Wyling's group where you

 

      could show mannitol, 2.3 grams of mannitol clearly

 

      had a big impact on bioavailability of cimetidine,

 

      a low permeability drug, and on the other hand,

 

      Fassihi had shown that a high permeability drug had

 

      minimal impact of sorbitol, even 10 grams of

 

      sorbitol.

 

                So, that was a hint, and we actually

 

      conducted a study at the University of Tennessee,

 

      and we have now completed that study, even getting

 

      to a mechanistic basis for generalizing the

 

      permeability effect to other excipients, is to test

 

      this out.

 

                Metoprolol was our boundary drug for high

 

      permeability, so we did a study with ranitidine and

 

      metoprolol, and we did a crossover study in

 

                                                                53

 

      replicate design to get an estimate of subject, the

 

      formulation interaction also.

 

                So, it is a very simple formulation.  You

 

      have your drug.  You have sucrose or sorbitol, and

 

      you have 15 ml of water.  That is the simplest

 

      study.

 

                What we found--I will just show you a

 

      picture--just a confirmation for a low permeability

 

      drug like ranitidine, a dramatic effect.

 

                Now we have completed the study of the

 

      dose response, the amount of sorbitol that triggers

 

      this is about 1.2 grams.  For metoprolol, the Cmax

 

      was affected, but not to the same extent.  AUC was

 

      not.

 

                In addition, there were a tremendous

 

      amount of information coming out of other excipient

 

      effects on transporters, and so forth.  So, this

 

      was an evolving issue at that time, and it

 

      continues to be evolving issue, and methodologies

 

                                                                54

 

      were being proposed of in vitro evaluation of this.

 

      I won't get into that.

 

                But we also did an extensive evaluation of

 

      excipients and what we found was I think in most

 

      cases, excipients that are used in solid dosage

 

      form really do not have a significant impact, but

 

      the way we had to evaluate that was comparing the

 

      differences in formulation that we have approved,

 

      like, for example, verapamil, and so forth.

 

                But the risk factor was excipients.  Is

 

      the current approach for evaluating excipients for

 

      decisions related to biowaiver of oral solutions

 

      sufficient?  Well, I think we left the question

 

      there.

 

                For BCS-based biowaivers, a higher

 

      standard was adopted by limiting biowaivers to

 

      highly permeable drugs. Excipients used in solid

 

      oral products are less likely to impact drug

 

      absorption compared to liquid oral products,

 

      because it was simply the volume and amount in

 

      there.

 

                For example, we had products on the market

 

                                                                55

 

      that contains 23 grams of sorbitol in one dose, so

 

      you can see cross interaction possibly.

 

                High permeability attribute reduces the

 

      risk of bio-in-equivalence, decreased small

 

      intestinal residence time by osmotic pressure,

 

      because low permeability generally have a tendency

 

      to be absorbed in small segments of the intestine.

 

                Clearly, on the other hand, the boundary

 

      that we chose for high permeability to be 90,

 

      because there are other surfactants, and so forth,

 

      that could increase permeability, so if you set

 

      your boundary at 90, there is no risk of failing.

 

                There were other examples.  There were so

 

      many examples that we had not really paid attention

 

      to.  Here is a submission--not a submission--this

 

      is a graph that a student of mine sent me from a

 

      company, and they have seen such effect.

 

                Here is a drug, a tablet and a solution.

 

      The solution has almost half the bioavailability of

 

      a tablet, so you can see that, with sorbitol or

 

      mannitol for a low permeability drug, it can have

 

      lower bioavailability.

 

                So, that was the basis that we came up

 

      with the recommendations and the boundaries for the

 

      BCS classification system that gave the basis for

 

                                                                56

 

      waiver for new drug applications.

 

                So, Class I drug, you have jejunal

 

      permeability. This was our research that we

 

      classified the number of drugs, and the volume of

 

      water required to dissolve the dose on the x axis.

 

      Class I dissolution in vivo is not likely to be

 

      rate limiting, and well characterized excipients.

 

      So, dissolution itself is likely to be rapid

 

      inherently, and then we can rely on in vitro

 

      dissolution for that purpose.

 

                Class II dissolution is likely to be rate

 

      determining and complex in vivo dissolution, and

 

      solubilization process, so no, not going there.

 

                Class III was where the debate was.  Some

 

      hesitation with the use of current dissolution

 

      test, because the site specific absorption was a

 

      concern, and excipients.

 

                Class IV was generally problem drugs with

 

      in vitro dissolution may not be reliable.

 

                So, that was the basis for our

 

      recommendations.

 

                So, wrapping up, in terms of quality

 

      design thinking, what can we do now?  If I

 

      summarize my concerns, one major concern was if we

 

      went towards a dissolution-based method, people

 

                                                                57

 

      will design products for dissolution rather than

 

      the intended use was a concern, and that example

 

      sort of illustrates that, the one to-be-marketed

 

      difference, that was a concern.

 

                So, I wanted to feel comfortable having

 

      some formulation assessment as part of this

 

      extension.  Clearly, I think excipients and the

 

      transporters, all were evolving issues at that

 

      time.  There is a lot more information available

 

      now than we had.

 

                So, in quality-by-design thinking, you

 

      really have focus on what are the critical

 

      variables that affect dissolution, and these are

 

      easily identifiable especially for immediate

 

      release dosage form.

 

                You easily can start thinking about

 

                                                                58

 

      excipients and what their impact might be on

 

      solubilization, and so forth, and Lawrence himself

 

      has done some work in this area, and so forth.

 

                So, with that in mind, what my thoughts

 

      are, in addition to thinking about evaluating the

 

      boundaries themselves, I would like to recommend to

 

      the group that, first of all, BCS should be, and

 

      probably will be, a key, too, in quality-by-design

 

      decision trees that we talked about.

 

                I mean solubility, permeability

 

      characterization has to be a starting point for the

 

      formulation, so clearly, I think we need to build

 

      these concepts in the decision trees we talked to

 

      you about yesterday, but also quality-by-design and

 

      design space with respect to pre- and post-approval

 

      by bridging studies.  Waivers for in vivo studies

 

      based on design space concept, sort of is that

 

      connection to extension concept.

 

                The challenge I think is from a generic

 

      industry perspective, there is a lot of hesitation

 

      to seek for approval, the first time approval, a

 

      biowaiver based on that, concerned with

 

                                                                59

 

      permeability assessment, and so forth.

 

      That concern probably will remain.

 

                It is not a scientific concern, it is a

 

      perceptional concern, it's a regulatory concern,

 

      and so forth, but that doesn't say that you cannot

 

      classify a drug for a generic product after

 

      approval.  The rest of the post-approval changes,

 

      they could be based on that, and that could be

 

      quality by design.

 

                I think those are the key connections that

 

      if the Technical Committee sort of starts building

 

      in, their efforts really get connected to the PAT

 

      and the quality-by-design thinking, so you see the

 

      connections sort of evolved.

 

                Clearly, they have already started

 

      developing in FDA's knowledge base, a knowledge

 

      base.  Drug-excipient interaction, I think is an

 

      increasing issue from chemistry and clinical

 

      pharmacology aspect, and I think you need to start

 

      connecting those dots.

 

                At the same time, drug substance and

 

      formulation variables and clinical performance that

 

                                                                60

 

      Mehul alluded to the variability, but with what is

 

      happening on the clinical side now, with focus on

 

      biomarkers, focus on surrogate markers, and so

 

      forth, I think we need to proactively keep an eye

 

      on that.

 

                I mean this is an evolving issue, but seek

 

      out some connectivity between quality and clinical,

 

      and be available to what is happening at least, and

 

      that is the point I made also to Jurgen yesterday

 

      when he gave his report.

 

                So, with that, let me stop and open it for

 

      discussion.

 

                DR. COONEY:  Thank you, Ajaz.

 

                Comments and questions from the Committee?

 

                DR. MEYER:  While I am formulating my

 

      question, I will ask another one, so I will give

 

      you some time to think.

 

                How confident are you that our knowledge

 

      base on excipients allows a reviewer to sit back

 

      and say, well, it has X, Y, and Z, and therefore,

 

      there is no problem?

 

                DR. HUSSAIN:  Well, I think the

 

                                                                61

 

      traditional excipients for immediate release dosage

 

      forms that we use as formulation aids for process

 

      ability, and so forth, I am fairly confident that

 

      there are very little concern there.

 

                There are other excipients that are

 

      necessary to aid in the dissolution process, such

 

      as surfactants and other aspect.  I think a close

 

      grouping of those would be necessary.  I think if

 

      we collect this information, it will start making

 

      the case, but if you have properties, such as high

 

      solubility, and so forth, you probably would not

 

      need those anyway.

 

                So, I think you can carve out excipients

 

      that we know are not an issue.

 

                DR. COONEY:  If I can just pick up on

 

      Marvin's point for a moment, your initial question

 

      was about knowledge base of the excipients per se,

 

      but it is really the relationship of the excipients

 

      to the drug substance, to the API, that seems to be

 

      the area of uncertainty, and that knowledge, it

 

      seems to me, is much less clear.

 

                DR. HUSSAIN:  If I amy sort of put that in

 

                                                                62

 

      the context, traditional screening experiments that

 

      are done for drug excipient compatibility, may

 

      provide not only information that will be useful

 

      for stability, failure modes of the product, but

 

      also hints about the interactions among excipients

 

      and drugs, how they might have a bearing on

 

      dissolution.

 

                DR. COONEY:  Ken.

 

                DR. MORRIS:  The point you just raised is

 

      the source of my question, as well, since

 

      drug-excipient interactions are typically for

 

      chemical stability.

 

                Is it like particularly for something like

 

      BCS Class III, is it more of the concern that the

 

      interaction with the drug is changing absorption,

 

      or that the interaction of the excipient with the

 

      mucosa or the sites of absorption?

 

                DR. HUSSAIN:  I think they are both

 

      concerns in the sense, but the concerns we had when

 

      we were working on this were more on the impact on

 

      the GI membrane, transporters, and so forth, the

 

      concerns with excipient-drug interactions that

 

                                                                63

 

      might be physicochemical were less of a concern,

 

      because we did not really focus on that aspect.

 

                DR. MORRIS:  That is sort of where I would

 

      assume it, but I just don't know enough biopharm.

 

                DR. HUSSAIN:  The aspect I think is this.

 

      I think the draft guidance that we have issued on

 

      polymorphism, for example, I think is a

 

      concentration there, in the sense what we have said

 

      is you could have a different polymorph, but if you

 

      can design your product well and if it meets the

 

      criteria, it's fine.

 

                So, I think that is the flexibility that

 

      already sort of comes through that, is that ability

 

      to demonstrate that, you can be different, but yet

 

      meet the intended use.

 

                DR. COONEY:  Art, then Marvin.

 

                DR. KIBBE:  This is more complex than we

 

      can handle in one or two days.  The number of

 

      excipients you use exponentially increases the

 

      possibility that one excipient is reacting with

 

      another excipient, that is reacting with the API.

 

                On top of that, the processing of the

 

                                                                64

 

      product changes things.  I mean we know all sorts

 

      of problems with mag. stearate and overblending,

 

      but we know certain issues. The question that

 

      always sits in the back of my mind is we have been

 

      discovering these issues on a regular basis over

 

      the last 20 years, have we discovered them all, and

 

      how can I be naive enough to think I have

 

      discovered them all, and I don't think I have.

 

                So, that gives me a basic uncomfort level

 

      with just waiving stuff when I really want a part

 

      that works in my patients.  So, I am uncomfortable

 

      with that.

 

                I have a question, a substantive question.

 

      If I make a soft gelatin capsule which contains a

 

      solution, is it a solution or is it a capsule?

 

                DR. HUSSAIN:  Capsule.  That's the way it

 

      is.

 

                DR. KIBBE:  On your basic I guess third

 

      slide, I could argue it's solution.

 

                DR. HUSSAIN:  Yes.  That's the aspect, I

 

      think now we can start thinking about those aspects

 

      if you have not done so in the past.

 

                But let me go back to the concern you

 

      raised in the sense, impact of magnesium stearate,

 

      and on dissolution, it is clearly documented.

 

                                                                65

 

      Impact of magnesium stearate on in vivo absorption

 

      has not been done yet.

 

                All the studies we have done, we had no

 

      impact of magnesium stearate on immediate release,

 

      and so forth, on in vivo absorption.  I could not

 

      find a single paper that conclusively tells me that

 

      what we see in vitro, the big difference is

 

      translating in vivo differences.

 

                There are two reasons for that.  One, is

 

      that old study that was published in 1967, J. Pharm

 

      Science, by Professor Newton from the University of

 

      London, where he demonstrated for lithium carbonate

 

      that if you include a very, very small amount, 0.01

 

      percent or 0.001 percent of sodium sulfate in your

 

      formulation, you negate the effect of magnesium

 

      stearate that you see in the solution.

 

                So, that was a hint to me that suggested

 

      that the surface tension differences that we see

 

      between in vivo fluids and in vitro fluids probably

 

                                                                66

 

      are the reason, because all the studies we did at

 

      the University of Maryland, we actually probed this

 

      for a low solubility drug--I am forgetting the name

 

      of the drug--we didn't see in vivo relevance of

 

      that.

 

                So, now that is the reason we have to

 

      start thinking about is risk-based decision to

 

      really understand the behavior of things in vitro,

 

      because one of the concerns that we had earlier was

 

      you see big differences in vitro, how do you know

 

      this will or will not translate.

 

                If quality-by-design, we are thinking, why

 

      is this assessment, then, that provides a basis to

 

      think about it.

 

                DR. KIBBE:  But that argument, I think

 

      would logically lead us to the conclusion that we

 

      have to go to a bioavailability study, we have to

 

      go to a clinical trial.  We can't rely on any of

 

      the standard tests that we do that are surrogates,

 

      because they don't work out, because they either

 

      show a problem that isn't real, or they ignore a

 

      problem that is real.

 

                DR. HUSSAIN:  Right.  That second bullet,

 

      that is what I am really thinking about.  If you

 

      have to qualify your design space, your

 

                                                                67

 

      bioavailability studies, if you are a new drug

 

      applicant, that becomes your test of hypothesis is

 

      to say that we have looked at these are the big

 

      differences we see that has an impact.

 

                So, one category of BCS-based biowaivers

 

      would be SUPAC related where you have demonstrated

 

      this in vivo, and that becomes the basis for that,

 

      and not just rely on in vitro testing and lack of

 

      that information.

 

                So, waiver is an extension of SUPAC in

 

      terms of design space is a bigger opportunity

 

      probably in the quality-by-design thinking.

 

                DR. COONEY:  Marvin.

 

                DR. MEYER:  Ajaz, I think I asked this

 

      question yesterday, and I think you said you were

 

      going to address it and maybe you did and I missed

 

      it, the rigid fixing of the--

 

                DR. HUSSAIN:  Boundaries.

 

                DR. MEYER:  --boundaries.  I really don't

 

                                                                68

 

      have any problem with the rigid definition of high

 

      solubility, high permeability, I mean we have

 

      pretty well nailed that down, but then you have, of

 

      course, if it's soluble in greater or less than or

 

      equal to 250, well, if it's only soluble in 300, is

 

      that really poor solubility, and if it's only 89

 

      percent absorbed, is that really low permeability,

 

      and, if so, does it fall in that 30 percent

 

      probability of failing a product?

 

                How do you deal with--you have to draw the

 

      line, but on the other hand, you draw the line, it

 

      becomes somewhat arbitrary and capricious.

 

                DR. HUSSAIN:  Very good point.  This is I

 

      think an important point because the objective of

 

      this guidance was to make the decision.  This is

 

      the decision.  You meet this, there is no issue.

 

      If you don't meet, you always have an option to

 

      explain, but nobody uses that option.

 

                So, this, in my opinion, is an approach

 

      that we had before.  In the new paradigm, the

 

      decision trees that we developed opens the door in

 

      a sense.  Here, the decision is pre-made, but

 

                                                                69

 

      instead of premaking that decision, how can you

 

      allow your science to drive a decision process that

 

      can justify the recommendations that come, but that

 

      then becomes specific to a company.  It is not a

 

      general guidance.  It is a decision tree to arrive

 

      at a proper decision.

 

                So, that would be an extension concept for

 

      BCS, not a general decision recommendation, which

 

      is what we have been trying to achieve.  It has

 

      changed the boundaries, and so forth.  But to

 

      incorporate that as a decision process, it becomes

 

      demonstrate this, and the decision can be yours

 

      sort of a thing.

 

                DR. MEYER:  So, it's sort of a work in

 

      progress, so to speak.

 

                DR. HUSSAIN:  It's not.  I mean I think

 

      the group has been busy with a number of things,

 

      but this isn't a thing that they could start

 

      thinking about, we have not done so.

 

                DR. COONEY:  Ajaz, if I can get clarity on

 

      that point.  I think the point you just made is

 

      that the decisions on class membership will be

 

                                                                70

 

      integrated into the thinking about the decision

 

      trees.

 

                DR. HUSSAIN:  Yes.

 

                DR. COONEY:  Is that correct?

 

                DR. HUSSAIN:  Definitely.  If you are a

 

      Class I drug and you exhibit the rapid dissolution

 

      with the conditions we have outlined, the decision

 

      is okay.  Anything else, the guidance does not

 

      recommend a waiver.  That's how it is.

 

                Based on what Marvin just suggested, and

 

      what I am formulating that as, this is a decision

 

      for every sponsor right now.  Their design and

 

      process understanding would vary from one end to

 

      the other end, but one way of extending this

 

      concept is not a general decision that this is

 

      where you get the waiver, but to define a decision

 

      tree and how you demonstrate to the degree of

 

      confidence that we need, that waiver would be you

 

      have demonstrated an understanding that the waiver

 

      will be granted.

 

                That will be Class I, that will be Class

 

      II, that will be Class III, so you have different

 

                                                                71

 

      levels of complexity in those, but the signs and

 

      the level of understanding could drive you to that,

 

      but that probably will become a post-approval as

 

      part of the design space.

 

                DR. COONEY:  Tom.

 

                DR. LAYLOFF:  I think I am not confused,

 

      but I don't understand some things like when a drug

 

      goes into the intestinal space, it is bound, not by

 

      water, but probably proteins and various other

 

      things that are present in the medium, and then it

 

      is absorbed through different sites depending on

 

      how it is wrapped in with the rest of the medium,

 

      and that is a drug-specific property, which then

 

      can be affected by an excipient, which might change

 

      the transport site, it may change the structure of

 

      the solution characteristics, is that correct?

 

                DR. HUSSAIN:  No, I think the basic

 

      premise is this, yes, you can have binding, you can

 

      have a number of other complexation reactions, and

 

      so forth, that occur.  Many of those are ionic and

 

      loose, so you establish equilibrium.

 

                For some drugs, you have complexation that

 

                                                                72

 

      really are almost very tight like with calcium and

 

      tetracycline, and so forth, there are a few such

 

      examples, but in this scenario, what we are talking

 

      about are equivalent behavior of the same drug

 

      molecule in two different formulations.

 

                So, if there are intrinsic properties of

 

      the drug molecule itself that will contribute, but

 

      that molecule is the same, that we are dealing with

 

      two formulations.  Now, how do formulations act

 

      with that behavior is a concern.

 

                I will sort of extend that concern to a

 

      paper that we had, a poster that we had, is that of

 

      precipitation.  A weak basis will dissolve very

 

      rapidly in acid conditions, but when they get

 

      emptied, there is a potential for precipitation,

 

      and so forth, and that could be a very complex

 

      process, and the size of the particles, not

 

      precipitation, crystallization may differ based on

 

      the excipients you have and the conditions you

 

      have.

 

                There is a potential that excipients could

 

      impact that.  So, that is generally Class II drugs,

 

                                                                73

 

      that's the boundary for Class I for high solubility

 

      was intended to prevent some of those things from

 

      occurring, too.

 

                DR. LAYLOFF:  Do you think the

 

      complexation and coordination around the drug

 

      substance would actually affect the transport site,

 

      change the site of transport, would change the

 

      properties of the system?

 

                DR. HUSSAIN:  Yes, it is clearly possible,

 

      but unlikely for an immediate release dosage form

 

      with the type of excipients we use.

 

                DR. MORRIS:  You know when I think about,

 

      it sort of makes my head hurt, but when I think

 

      about--

 

                DR. HUSSAIN:  It's complex.

 

                DR. MORRIS:  --the amount of time we spent

 

      working on developing design spaces for the

 

      processing end of things, which as Jerry says, may

 

      be a way off, but still in comparison relatively

 

      simple to the larger problem, is there an

 

      opportunity in the context of using development

 

      data to somehow leverage tox studies to be able to

 

                                                                74

 

      get early reads on, not the tox itself, but in

 

      terms of some of the dynamics that are going on

 

      with the dosage forms?

 

                DR. HUSSAIN:  I have a decision tree,

 

      which I did not present yesterday, but it was part

 

      of the handout.  That decision tree came out of our

 

      AAPS workshop on how to leverage that.  The paper

 

      is published, Diane Burgess [ph], Eric Duffy from

 

      FDA is on it, so it is there in your handout.  I

 

      don't have it in this one.  Take a look at it.

 

                That leverage is every bit of information

 

      coming from Pharm Tox, and so forth, to start

 

      building that case for that.

 

                DR. MORRIS:  For the design of the dosage

 

      form?

 

                DR. HUSSAIN:  Yes, for particle size

 

      dissolution and bioavailability concentration.

 

                DR. COONEY:  Marvin.

 

                DR. MEYER:  Ajaz, when a generic company

 

      sends in their ANDA, it was my understanding that

 

      the generic group does not go back to the NDA to

 

      review the contents of the NDA, so they don't look

 

                                                                75

 

      at the excipients and see which excipients are now

 

      different in the ANDA than were in the NDA product?

 

                DR. HUSSAIN:  I will let Lawrence answer

 

      that, but we do have a process of inactive

 

      ingredient guide that we consult, and so forth.  I

 

      put him on the spot here.

 

                DR. YU:  I guess this morning we talk

 

      about excipients, which emphasize how complex the

 

      process is.  Yes, with advances in molecular

 

      biology, we discovered I even don't know how many

 

      transporters going on.  As far as the PGP, at least

 

      32 and 64 is transporters, however, I want to

 

      emphasize that how those transporters impact

 

      absorption we rarely see in clinical settings.

 

                In other words, we very see excipients

 

      impact on absorption of Drug A, B, C, D, but in

 

      vivo setting, we have very, very few, two or three

 

      publications out there compared to tens of

 

      thousands of publications to show that excipients

 

      impact in vitro.

 

                So, I have to say that we still want to

 

      see more evidence to show the impact of excipients

 

                                                                76

 

      on absorption of drugs in general.

 

                Secondly, while we see the impact exceeds

 

      absorption, we very open to see the unique of some

 

      of the products out there.  The reality for, say,

 

      70 or 60 percent of immediate release products,

 

      people espouse the intensity, use very limited

 

      number of excipients, I would say 10, within 10.

 

      For example, Avacel almost uses the majority of

 

      products.  All those excipients impact, and have

 

      not seen in vitro, as well as in vivo.

 

                Certain, because of those common used

 

      excipients, since we have a sufficient knowledge to

 

      judge whether they are going to impact absorption

 

      or not, will generate and not see the formulations,

 

      however, in very few cases, some cases, we suspect

 

      potential impact of excipients absorption, we will

 

      look into it further before we make any scientific

 

      decisions about approvability of any NDAs.

 

                I hope that answers his question.

 

                DR. HUSSAIN:  The other aspect, just to

 

      build on what Lawrence said, traditionally, the

 

      composition, especially for immediate release, it

 

                                                                77

 

      is hardly any different than the quality.

 

                DR. MEYER:  My real question was do you go

 

      back and look at the NDA to see if Pfizer used

 

      Avacel, and Teva used who knows what, do you make

 

      that comparison, say, well, wait a minute, they are

 

      putting in two things instead of Pfizer's one

 

      thing.

 

                Can that potentially make a difference?

 

      Do you review the NDA product composition?

 

                DR. YU:  Well, certainly, we will review

 

      any scientific literature out there and information

 

      available to us to make the best decisions.

 

                DR. MEYER:  But do you review the NDA?

 

                DR. HUSSAIN:  Marvin, often we don't have

 

      to, because it's in the label.

 

                DR. MEYER:  Well, that's true.  It didn't

 

      used to be.

 

                DR. HUSSAIN:  But definitely, the criteria

 

      there is to look at what has been approved and what

 

      has been used in dosage forms and inactive

 

      ingredient guide, and so forth.

 

                DR. YU:  I guess the answer is as long as

 

                                                                78

 

      are trying to build in the science base or any ANDA

 

      decisions,

 

      we will use any information which is available,

 

      whether scientific literature or not, to us.

 

                DR. COONEY:  Are there any other questions

 

      at the moment?  Thank you, Ajaz.

 

                We are running a bit ahead of schedule.  I

 

      think this would an appropriate time to take a

 

      break for 15 minutes.  We will reconvene at 8 past

 

      10:00 and then begin immediately with Lawrence Yu's

 

      presentation.

 

                [Break.]

 

                DR. COONEY:  I appreciate everyone's

 

      diligence to staying on time.  It has worked very

 

      well.

 

                Lawrence Yu will proceed with a

 

      presentation on Using Product Development

 

      Information to Address the Challenge of Highly

 

      Variable Drugs.

 

                Lawrence.

 

            Using Product Development Information to Address

 

                 the Challenge of Highly Variable Drugs

 

                DR. YU:  The assignment to me today, this

 

      morning, is for me to discuss how to use

 

      pharmaceutical development information to address

 

                                                                79

 

      or potentially address the bioequivalence issues of

 

      highly variable drugs.

 

                Before I go on and talk about how to use

 

      or potentially use the pharmaceutical development

 

      information to highly variable drugs by equivalency

 

      issues, I want to give you a very brief overview or

 

      update what has been happening before.

 

                For highly variable drugs, this is not the

 

      first time, it's the second time we present it to

 

      you.  In the first presentation on April 14th of

 

      2004, we discussed the challenges and the

 

      opportunities for bioequivalence of highly variable

 

      drugs.

 

                At this meeting, the objective was to

 

      explore and define bioequivalence issues of highly

 

      variable drugs, for example, what is called highly

 

      variable drugs and discuss potential solutions to

 

      deal with the bioequivalence of highly variable

 

      drugs.

 

                We invited a number of speakers from

 

      industry, academia to address issues related to

 

      bioequivalence including why the bioequivalence of

 

      highly variable drugs is an issue, highly variable

 

      drugs a source of variability by Gordon Amidon from

 

      the University of Michigan, and the clinical

 

                                                                80

 

      implications of highly variable drugs by Leslie

 

      Benet, and from bioequivalence method include the

 

      skin method by Laszlo, as well as bioequivalence of

 

      highly variable drugs, we had a good discussion at

 

      this meeting.

 

                I want to highlight some of the things

 

      which have been discussed at this meeting,

 

      particularly the slides by Professor Leslie Benet

 

      from the USCSF.  His talk with implications of

 

      highly variable drugs, the argument was why highly

 

      variable drugs are safer.

 

                Specifically, he said for wide therapeutic

 

      index highly variable drugs, we should not have to

 

      study the excessive number of patients to confirm,

 

      to demonstrate that two equivalent products meet

 

      the preset statistical criteria or by equivalent

 

                                                                81

 

      standards.

 

                This is because, by definition, highly

 

      variable approved drugs must have a wide

 

      therapeutic index, otherwise, there have been

 

      significant safety issues and lack of efficacy

 

      during Phase III.

 

                Highly variable narrow therapeutic index

 

      drugs are dropped in Phase II since it is

 

      impossible to prove either efficacy or safety.

 

                Now, for the benefit of some new members

 

      for this committee, I have two slides to briefly

 

      review why this issue, why the one-size-fits-all,

 

      what we are using today.

 

                In order to determine bioequivalence, we

 

      normally define as a rate of bioavailability,

 

      defined as a rate and extent of drug absorption.

 

      Bioequivalence is defined as absence of significant

 

      difference in the rate and extent absorption.

 

                In practice, when we give the drugs

 

      orally, for example, to a healthy volunteer, we

 

      will draw the blood.  We got the plasma

 

      concentration profile.  We are certainly not able

 

                                                                82

 

      to get exactly how much and how fast drug gets

 

      absorbed, therefore, in practice, we use AUC, area

 

      under the curve, to represent extent of absorption.

 

                We use Cmax as a surrogate for the rate of

 

      absorption, certainly in some cases we also look at

 

      Tmax, because indeed, if you look at Tmax and Cmax

 

      here, it represents the rate of absorption.

 

                So, from that, we will define what the

 

      bioequivalence study is passing or not passing.

 

      Basically, the bioequivalence criteria, either

 

      statistical criteria here is 80 to 125 percent.

 

                At this date, that is the

 

      one-size-fits-all regardless drug, drug product,

 

      regardless of therapeutic class, regardless for

 

      anything, that bioequivalence study, you have to

 

      use preset, so-called bioequivalence criteria,

 

      which is 80 to 125 percent.

 

                Now, let's look and explain why the highly

 

      variable drug is an issue.  Let's look at the red

 

      one.  If you use a highly variable or intersubject

 

      variability is high.  Statistical confidence

 

      interval, if you use the same number of subjects,

 

                                                                83

 

      when variability goes higher, the confidence is

 

      going wider.  When confidence gets wider, it

 

      becomes more and more difficult pass the confidence

 

      interval or bioequivalence interval if 80 to 125

 

      percent.

 

                So, that explains when the variability

 

      goes higher, it gets more and more difficult to

 

      pass a study.

 

                On the other side, in order to narrow the

 

      confidence interval, for example, here it is fair

 

      to demonstrate bioequivalence for super red one

 

      here in order to make confidence interval narrower,

 

      you have to use a large number of subjects, because

 

      the higher the variability, the higher the

 

      confidence interval, the higher the number of

 

      subjects in general, the narrower the confidence

 

      interval.

 

                Therefore, for highly variable drugs, you

 

      will have to use higher number of subjects.  Just

 

      to give you example here, for example, normally, we

 

      have a 20 percent or 30 percent intersubject

 

      variability.  You maybe use 18, 24, even sometimes

 

                                                                84

 

      for good product or good drugs, you only need to

 

      use 12, actually, they can pass the bioequivalence

 

      confidence interval.

 

                But this is not always true, because when

 

      the variability goes higher, now, this variability

 

      could be because of a drug, or it could be because

 

      of a product, so think about if variability goes

 

      100 percent--some of you think 100 percent, that is

 

      unrealistic, but we do have a drug, we do have

 

      examples--think about with 100 percent variability,

 

      assume test and the reference, there is 5 percent

 

      difference, you end up it could be 500 or more

 

      subjects, or 300 subjects, so this is certainly a

 

      large number of subjects in order to pass the

 

      bioequivalence study.

 

                So, Leslie argued at the previous meeting,

 

      from the clinical perspective, this is not

 

      necessary.  To give you a real example, these are

 

      slides from Leslie Benet.  Now, you would argue,

 

      you may ask how do we get intersubject variability.

 

                Certainly, you could get this number from

 

      literature or sometimes company conduct a pilot

 

                                                                85

 

      study, get some kind of estimate how many subjects

 

      need to be used to pass the bioequivalence study.

 

                Of course, in this case, based on

 

      intersubject variability, you need to use 300, now

 

      this is the drug.

 

                So, at the previous meeting, when we

 

      present the issue to you, and also we present some

 

      of the possible potential solutions including

 

      widening the confidence interval.  Now, that is

 

      very straightforward.  You said by confidence 80 to

 

      125 is too narrow for highly variable drugs, and

 

      your intuitive thinking is just widening the

 

      confidence interval, that is one of the potential

 

      solutions.

 

                Another solution is a scaling approach, in

 

      other words, based on the variability of reference

 

      list product, reference list drugs, and calculate,

 

      use statistical approaches to calculate the

 

      confidence interval, then, to determine whether the

 

      study is passing or not.

 

                Obviously, I have to say this.  The active

 

      approach because the confidence interval too wide

 

                                                                86

 

      in order for the study to pass, so let's widening

 

      the standards.

 

                You came in and asked to do that, that

 

      certainly the scaling approach, we ought to

 

      carefully look into, the Committee suggested--a

 

      quote here--"the need to understanding where the

 

      variability originated.  The members added that

 

      prior knowledge of all biostudies may help set more

 

      appropriate specifications or criteria to make

 

      decisions.

 

                So, you suggest that we have to understand

 

      the origin of the variability.  Now, to look at the

 

      mechanistic understanding of variability for drug

 

      substance obviously is the same, reference list

 

      product and the generic product, or any other

 

      product, but the potential difference could be

 

      formulation.  Certainly, the generic products could

 

      be narrower or could be wider, the variability.

 

                We believe at this point, in order to

 

      understand the origin of the variability, that

 

      pharmaceutical development report, or

 

      pharmaceutical development information can help us

 

                                                                87

 

      understand the source of variability, can help us

 

      make rigid scientific evaluation.

 

                Now, in order to see the utility of

 

      pharmaceutical development report to evaluation or

 

      reviews of NDAs, let me go back and review some of

 

      the basic fundamentals or the premises for ANDA

 

      approvals.

 

                Ajaz has mentioned about therapeutic

 

      equivalence. Basically, the products are considered

 

      to be therapeutic equivalents only if they are

 

      approved as safe and effective, they are

 

      pharmaceutically equivalent, they are

 

      bioequivalent, adequately labeled, and manufactured

 

      according to cGMP.

 

                Now, here, I want to emphasize the

 

      pharmaceutical equivalence.  When we define

 

      pharmaceutical equivalence, we basically have the

 

      same active ingredients, obvious.  I know we are

 

      managing about pharmaceutical solid polymorphism,

 

      which was presented to you two years ago, has a

 

      drugs guidance out there and published by FDA in

 

      December of 2004.

 

                You have to be same dosage form, same

 

      route of administration, and identical in strength

 

      and concentration, and may differ the other

 

                                                                88

 

      characteristics, such as shape, excipients,

 

      packaging, and so on, and so forth.

 

                Now, under the same dosage form, I think

 

      what the complexity of dosage form is particularly.

 

      Yes, I would say several decades ago, that dosage

 

      form is reasonably simple and in most cases I would

 

      say the immediate release product or solutions.

 

                Certainly, with advances of pharmaceutic

 

      industry and the pharmaceutic technologies,

 

      so-called dosage form gets more and more complex,

 

      and we now have the soft gel capsules, we have

 

      ricin [?] product, we have inhersion [?] product,

 

      presents additional challenge to us in terms to

 

      make scientific decisions, in terms of make

 

      scientific evaluations.

 

                We therefore would believe a

 

      pharmaceutical development report,

 

      quality-by-design, designed to be equivalent,

 

      become more and more significant in this regard.

 

                Why does pharmaceutical equivalence

 

      matter?  Because of user experience and

 

      expectations.  Then, bioequivalence test is

 

      normally conducted in healthy subjects.  Certainly,

 

      we have assumption that equivalence in healthy

 

      subjects equals equivalence in patients.  Now, we

 

                                                                89

 

      have many, many generic products out there which

 

      are safe, which is a high quality, which are

 

      effective, which is the equivalent to the reference

 

      list product.

 

                So, certainly, we have tremendous

 

      experience with that, and certainly the

 

      pharmaceutical equivalence presents more and more,

 

      become because you want to make sure the data from

 

      the healthy volunteer does the equivalent in

 

      patient, and against novel drug delivery systems

 

      presents a challenge.

 

                That is why we want to use more and more

 

      pharmaceutical development approach to make a

 

      judgment, pharmaceutical development information to

 

      make a scientific judgment.

 

                Highly variable drugs very often have, as

 

                                                                90

 

      I mentioned, a wide therapeutic index, and the

 

      clinical trials of reference list product have

 

      established the acceptable level of variability,

 

      because I said otherwise, these highly variable

 

      drugs, a big window index, they will be dropped in

 

      Phase II.

 

                So, under an ideal situation, you will

 

      think about variabilities very high, so you will

 

      think it should be easier to pass, easier to design

 

      equivalent product simply because they are so wide,

 

      the target is wide, so it is easier for you to

 

      pass.

 

                Obviously, as I said before, if you use

 

      the preset 80 to 125 bioequivalence confidence

 

      interval, it is not the case.  While we explore our

 

      tentative approaches to deal with the

 

      bioequivalency issues, certainly, the design issues

 

      come out.

 

                So, now, how do we deal with

 

      pharmaceutical development for highly variable

 

      drugs?  Obviously, sponsor need to understand what

 

      are reference products supposed to do with origin

 

                                                                91

 

      of variability, and the purpose of design can be

 

      equivalent, and to evaluate and to verify the

 

      design and hopefully, in the future, use the

 

      bioequivalent study design for highly variable

 

      drugs.

 

                So, we put more emphasis on design in this

 

      regard to establish pharmaceutical equivalence, in

 

      order to establish therapeutic equivalence, which

 

      will be more appropriate.

 

                While we are looking for shared

 

      information for generics with us, there is a reason

 

      for doing that, not only for evaluation for highly

 

      variable drugs, certainly for pharmaceutical

 

      development is required.  It's one of the CTD

 

      format.  It is also outlined in ICH Q8, although

 

      they do not apply to us, but I think some

 

      principles should apply to generic industry also.

 

                Also, more significantly, OGD question

 

      based on review.  Now, this is still a work in

 

      progress, but I want to share some questions, I

 

      think it is important to ask to share.

 

                What is the formulation intended to do? 

 

                                                                92

 

      What mechanism does it use to accomplish this?

 

      Were any other formulation alternatives

 

      investigated and how did they perform?  Is the

 

      formulation design consistent with the dosage form

 

      classification in the label?

 

                So, those questions will help us get

 

      information about a pharmaceutical product, the

 

      report will help us, pharmaceutical product design

 

      and development, make more sound and appropriate

 

      scientific determination or evaluation.

 

                The question often comes up, why do we

 

      need to provide those things to the FDA?  That

 

      again is a quality-by-design paradigm, and

 

      pharmaceutical development report is where you

 

      demonstrate the drug is highly variable.

 

                Now, this demonstrate not necessarily to

 

      study, but you certainly use any source available

 

      to show why this drug or drug product is highly

 

      variable, and may use a different criteria other

 

      than 80 to 125 percent confidence interval.

 

                Also, the pharmaceutical development is

 

      where you justify equivalence of design, why do you

 

                                                                93

 

      think the product which you designed is equivalent

 

      to the reference list product.

 

                During the discussion, the members ask

 

      whether it is drug or drug product.  Now, for

 

      example, Product A, the variability is the active

 

      ingredient into exceptions, so formulation design

 

      could be rapid release, so demonstrated by

 

      dissolution comparison under physiologically

 

      relevant conditions, if this is BCS Class I drug,

 

      which is highly soluble, highly permeable, even

 

      though they are highly variable, you may still

 

      require biowaiver, otherwise, you will have to

 

      conduct some bioequivalence studies to demonstrate

 

      that they are bioequivalent.

 

                Certainly, the approaches to deal with

 

      highly variable drugs, to deal with the

 

      bioequivalence of highly variable drugs are still

 

      in discussion and still in investigation.  I am

 

      hoping in the near future we share with you some

 

      proposal or recommendation we have with respect to

 

      bioequivalence of highly variable drugs.

 

                Another drug could be drug product, the

 

                                                                94

 

      drug product could be highly variable, even drug

 

      substance is I would say low variability, and

 

      certainly design for equivalence begins with the

 

      characterization of the reference list product, and

 

      generic product should target the mean, and the

 

      current system again would have no reward for

 

      narrow or less variability of generic products.

 

      That is why we need to explore the alternative

 

      approaches or more appropriate approaches to deal

 

      with highly variable drug products.

 

                I just want to give you some examples of

 

      what we talk about here.  This is real data.  This

 

      is single subject replicate design, in other words,

 

      you give the same product to the same patient

 

      twice.  Here is the plasma level, obviously, I am

 

      sure that out of 80 to 125 percent confidence

 

      interval, by any standards, it probably does not

 

      need a statistician to figure this out.

 

                You can see here, this is the first period

 

      or this is the second period.  It is not in your

 

      handout or printout because this is in color.  If

 

      you look carefully, these two curves are

 

                                                                95

 

      significantly different, probably different by I

 

      even don't know how many folders.

 

                This is a single-dose study twice,

 

      replicate study design.  Sorry, I should do a

 

      better job next time, use red, so you can see it.

 

                DR. KIBBE:  I am just looking at the curve

 

      and wondering why we got the hump at the back end

 

      and whether the product is intended to have a

 

      second release.

 

                DR. YU:  No, it's simply by design, for

 

      whatever reason this peak has come out.  Obviously,

 

      a second dose, this peak is no longer there.  So,

 

      it's not purpose designed, it's simply because of

 

      physiology involved.

 

                This is happening because enteric coated,

 

      this is coated to release at the target pH, so when

 

      the physiological pH in the gastrointestinal tract

 

      may fluctuate, and those curves will change.

 

                Think about, for example, if we have a

 

      product designed will release a pH 7, so then in

 

      the terminal ileum, at one point is pH 6.8, you

 

      will not see the release.  But a second day,

 

                                                                96

 

      because of food or because of other reasons,

 

      terminal ileum pH becomes 7 or 7.2, you do not see

 

      the release.  Otherwise, you will not see it.

 

                So, simply pH effect or significant impact

 

      the absorption.

 

                DR. KIBBE:  The product had gotten on the

 

      market because it worked clinically?

 

                DR. YU:  Yes.  Even though we see the

 

      significant variation in pharmacokinetics, but we

 

      have no reason to believe this variation will

 

      impact safety and efficacy.

 

                So, in order to do more appropriate

 

      pharmacokinetic studies, we also look into what

 

      additional information, for example, develop

 

      information will help support those cases or

 

      bioequivalence cases, because you can see the

 

      bioequivalence obviously is very difficult to

 

      conduct variability probably up to 2 or 100

 

      percent, and the number of subjects very high.

 

                So, we want to see can we use any

 

      additional pharmaceutical development information

 

      to help us to make more proper scientific

 

                                                                97

 

      decisions.

 

                Again, for example, when we are looking,

 

      in many cases, we do get very consistent, the in

 

      vitro dissolution actually out to say the majority

 

      of cases, those help us out to make a more proper

 

      scientific decision or rational scientific decision

 

      when we recommend any method to demonstrate

 

      bioequivalence, but occasionally, we do get very

 

      strange results, and actually, the variability is

 

      extremely high and does not help you.

 

                I just want to show you another case here

 

      when we conduct the dissolution under physiological

 

      relevant pH condition, and you get dissolution all

 

      over the place.

 

                Now, this is a 6 tablet, same lots, same

 

      bottle, put in 6 vessels, you get a distortion

 

      curve.

 

                So, the next question we ask, this is a

 

      large variability because of the operator or

 

      because of other reasons.  I think the answer is we

 

      are almost certain those difference is because of

 

      product, not because of other factors.

 

                So, what I can present to you today is we

 

      have challenges to deal with bioequivalence of

 

      highly variable drugs.  We use the clinical

 

                                                                98

 

      evaluation and sometimes we are also facing

 

      challenges when we are trying to use in vitro

 

      information to help us make decisions.

 

                DR. SINGPURWALLA:  Lawrence, what is the

 

      difference between each curve, different vessels?

 

                DR. YU:  Yes, six vessels.

 

                DR. SINGPURWALLA:  Six vessels, so it

 

      could be that the vessels are different.

 

                DR. YU:  I think I stated that the

 

      variability because other reasons, for example,

 

      vessel difference, media difference, degassing

 

      difference, operator difference, assay difference.

 

      We do not believe all these reasons can explain.

 

                DR. HUSSAIN:  Yesterday, this same figure,

 

      Cindy actually showed you the reason for this

 

      difference was the coating thickness, and so forth,

 

      so this is the same slide.

 

                DR. MORRIS:  You wouldn't get 2 1/2 hours

 

      difference in dissolution time from different

 

                                                                99

 

      vessels.  That is not the magnitude you would

 

      expect.

 

                DR. SINGPURWALLA:  How am I supposed to

 

      believe that?

 

                DR. YU:  You have to believe in me.  You

 

      don't have any other options.

 

                [Laughter.]

 

                DR. SINGPURWALLA:  I don't believe in

 

      anyone.

 

                DR. KIBBE:  This is a constant pH

 

      throughout, right, we haven't shifted pH during the

 

      process or anything, right?

 

                DR. YU:  Correct.

 

                DR. HUSSAIN:  The reason to believe that

 

      is I think it was done by Cindy, and with our

 

      stringent mechanical calibration.

 

                DR. LAYLOFF:  He demonstrated it with

 

      variable coating.  It's variable coating on enteric

 

      coating material, so if there is a crack in the

 

      coating, it disintegrates much more rapidly.

 

                DR. SINGPURWALLA:  I think I believe Tom.

 

                DR. YU:  Thank you very much.

 

                So, the objective, the case we presented

 

      to you is certainly difficult, I just want to say,

 

      variability issue, whether from clinical evaluation

 

                                                               100

 

      or sometimes for in vitro conditions, in vitro

 

      testing, target main performance question.  I am

 

      sure you will ask where is the main performance.

 

                I just want to show you that these are the

 

      challenges which we are facing, and certainly we

 

      are open to any suggestions or input from you.

 

                So, in summary, we believe pharmaceutical

 

      development information will help.  I quoted here,

 

      that's the conclusion made by you April 14th of

 

      2004.  Understanding what the problem is, as well

 

      as the real fundamentals, for example, physical and

 

      chemical parameters, and make coherent and

 

      scientific science-based decision based on

 

      pharmaceutical development information, I think I

 

      present to you the cases to see hopefully how we

 

      use pharmaceutical development information to help

 

      us in most cases, but in some cases, we still have

 

      challenges and we have opportunities for us to move

 

      forward.

 

                Thank you and any comments are welcome.

 

      Thank you very much.

 

                DR. COONEY:  Thank you, Lawrence.

 

                We now have time for questions from the

 

      Committee.

 

                DR. SINGPURWALLA:  Lawrence, I have two

 

                                                               101

 

      kinds of questions.  Question No. 1.  Is it the

 

      purpose of this presentation of yours to ask the

 

      manufacturers, namely, the industry, to provide

 

      more information to you because there is so much

 

      variability and you are trying to get to the source

 

      of the variability, is that the objective?

 

                DR. YU:  Yes, very precise, certainly much

 

      better than I said.

 

                DR. SINGPURWALLA:  That is the political

 

      question. The scientific question, and I have heard

 

      this before, what does T/R percent mean in your

 

      Slide No. 8?

 

                DR. MEYER:  Test over reference.

 

                DR. SINGPURWALLA:  Test over reference.

 

                DR. YU:  Yes.

 

                DR. SINGPURWALLA:  How was this 80 percent

 

                                                               102

 

      and 125 percent figure arrived at?

 

                DR. YU:  Slide 9.  I am trying to get

 

      Slide 9.

 

                DR. SINGPURWALLA:  That's it, the picture.

 

      So, 80 percent and 125.

 

                DR. YU:  T is the test.

 

                DR. SINGPURWALLA:  No, forget that.  How

 

      did you get 80 and 125?

 

                DR. YU:  That's an excellent question, and

 

      we have been asked many times.

 

                DR. SINGPURWALLA:  It can't be excellent.

 

                DR. YU:  It's back to it was published

 

      when I was in high school, I would say, 20 years

 

      ago, or even more than 20 years ago, when the

 

      pharmacokinetics, the discipline was developed, and

 

      FDA developed the criteria.  Actually, this

 

      evolving process and trying to develop what kind of

 

      standards or criteria can we use to judge a

 

      bioequivalence study is okay or is not okay.

 

                I think at that time, the physicians get

 

      together, as we do today, and the physicians

 

      together made the determination that the 20

 

                                                               103

 

      percent, the difference between product would not

 

      be considered clinically significant, because the

 

      20 percent will not be considered significant

 

      difference, therefore, when translated into in vivo

 

      setting, you have 80 percent.

 

                So, you would think from 80 to 120 instead

 

      of 25. Now, in the normal processing of

 

      pharmacokinetic data, they used log normal to be

 

      much better to describe the distribution.  So, when

 

      you use log normal, 80 is still 80. When you have

 

      the 1 over 80 or 1 divided by 0.80, equals 1.25.

 

      That is why you see 80 to 125.

 

                Now, at the beginning, I would think 20

 

      percent instead of 19 percent or 21 percent, which

 

      is 20 percent, it was decided.  Then, the question

 

      come back to us now can we change 20 percent to 25

 

      to 15, 10, 5 percent, and I guess we have to use,

 

      say, over the 20 or 25 years, we approved product,

 

      they are all safe, they are all equivalent, they

 

      are all high quality, because of those experience

 

      or prior knowledge, determining 80 to 125 percent

 

      works fine.

 

                Now, this does not necessarily mean we

 

      cannot change it, but the criteria we have is very

 

      stringent criteria, we feel confident with that.

 

                                                               104

 

                Now, with a statistical interplay--

 

                DR. HUSSAIN:  Lawrence, if I may.

 

                DR. YU:  Yes, please.

 

                DR. HUSSAIN:  It's a "feel good" criteria,

 

      we felt good about it.

 

                DR. SINGPURWALLA:  I got the answer.  I

 

      think I got the answer.

 

                [Laughter.]

 

                DR. SINGPURWALLA:  The answer is

 

      tradition.

 

                DR. HUSSAIN:  No, it's rational science.

 

                DR. YU:  It is rational science.  I think

 

      I proved it.

 

                DR. SINGPURWALLA:  Let me make a

 

      suggestion.

 

                DR. YU:  Yes, please.

 

                DR. SINGPURWALLA:  That tradition with

 

      some dose of rationality was good 20 years ago when

 

      you were in high school.  Times have changed. 

 

                                                               105

 

      These kind of decisions to either prove equivalence

 

      or prove in-equivalence should be based on risk

 

      considerations and should be based on appropriate

 

      utilities.

 

                So, I think it is time to change, and I

 

      think I said that April 14th, 2004.  Has there been

 

      any progress made towards changing?

 

                DR. YU:  The answer is yes.

 

                DR. SINGPURWALLA:  Oh, good.  What?

 

                DR. YU:  Certainly, you said you want

 

      suggestion of change, and I think under the

 

      leadership of Gary Buehler, that we are exploring

 

      the confidence interval, for example, the window

 

      index drugs, and also we are exploring confidence

 

      interval for highly variable drugs.  In other

 

      words, in the future, I am hoping someday, with

 

      your support and agreement, we will have different

 

      criteria other than 80 to 125 to different class of

 

      drug in consideration of the risk interplay.

 

                Obviously, to make any changes, six months

 

      or one year is not enough.

 

                DR. COONEY:  Marvin, then Ken, then Paul.

 

                DR. MEYER:  Your talk I believe tried to

 

      marry the quality-by-design to the highly variable

 

      drug and show that you could, in part, solve the

 

                                                               106

 

      problem by quality-by-design, that's the objective.

 

                DR. YU:  Yes.

 

                DR. MEYER:  Personally, I think if you

 

      have a competent company, then, your highly

 

      variable drug is biological problem which the

 

      company can't solve.  You have to speak directly to

 

      a higher power to get rid of that variability.

 

                So, I think, yes, there is cases where,

 

      for example, I could cite failure by design if you

 

      want to put an enteric coating on something,

 

      because that is, in my view, not a good dosage form

 

      because it is so dependent on gastric pH and

 

      emptying, and all of that, so you are setting

 

      yourself up for failure.

 

                Now, you can say, well, I dealt with

 

      quality by design by not using enteric coated, I

 

      kind of took the reverse of that.  A competent

 

      company looking at Slide 24, the 6-vessel graph,

 

      would never go to a biostudy with a product that

 

                                                               107

 

      showed dissolution characteristics like that.

 

                So, indeed, some quality built in that

 

      says whoa, let's not spend $100,000 on a biostudy

 

      when our drug is all over the map in dissolution.

 

      So, I think you can deal with some variability, but

 

      that is fairly straightforward I think for a

 

      company.

 

                So, the issue that really faces us is the

 

      physiological variability and do we extend the

 

      confidence limits, do we have point estimate

 

      restrictions or just do we do 600-subject studies.

 

                DR. HUSSAIN:  Marv, may I just sort of put

 

      that in context a bit?  In some ways, what we are

 

      seeing here is this.  Since we are comparing two

 

      formulations of the same drug, the drug is the

 

      same, the variability, the physiologic, the

 

      variability that is coming is the same for the drug

 

      substance.

 

                If we can compare formulations and say

 

      that all the conditions that are critical to

 

      exposure are well controlled, and so forth, and get

 

      confidence, what will give us the confidence to say

 

                                                               108

 

      that the inherent variability is the physiologic

 

      variability, not the quality variability, then, we

 

      can move forward.  I think that is the hope that we

 

      hope.

 

                DR. MEYER:  Do you think practically, you

 

      can look at the restrictions and the SOPs--

 

                DR. HUSSAIN:  No.

 

                DR. MEYER:  --and just see how a company

 

      is formulating and designing and developing a

 

      product?

 

                DR. HUSSAIN:  Not with the traditional

 

      work we do about formulations, putting things

 

      together, and so forth, no, it has to be a

 

      structured design approach that goes through

 

      identifying the sources of variability in your

 

      materials, and so forth, and putting a convincing

 

      case together to say based on the assessment, in

 

      this case it's a generic product, and based on

 

      characterization of reference material and your

 

      test product, you can make the case that the

 

      variability that you are seeing in your product is

 

      no more different than of the best argument.

 

                That gives you a leverage to now make a

 

      rational decision with respect to what sort of a

 

      biostudy criteria would be necessary.

 

                                                               109

 

                You can build flexibility, and not go

 

      rigid with, say, the Japanese approach, which was

 

      in your background packet, was to say do we really

 

      need confidence interval criteria here.  We just

 

      want to confirm the mean values.  It's a

 

      confirmation rather than a complete full-fledged

 

      study.  One option could be that.

 

                DR. YU:  I think the message we are trying

 

      to convey is when we explore alternative approach,

 

      which could be a wide confidence interval, or your

 

      scaling approach to show or to demonstrate the

 

      bioequivalence is demonstrated with the additional

 

      information, which is pharmaceutical development

 

      information, will help us to make scientific

 

      coherent decisions.

 

                Right now we don't, we don't have those

 

      informations.  In other words, we are not able to

 

      see how dissolution variability here may change it,

 

      for example, in this case, if we change the pH,

 

                                                               110

 

      dissolution is very beautiful, so that is the data

 

      we got.  We have now seen this data I showed you on

 

      the screen.  Thank you.

 

                DR. COONEY:  Ken.

 

                DR. MORRIS:  A couple of points.  One is I

 

      agree with Marvin in the sense that you wouldn't

 

      expect a company to release dissolution, I mean

 

      going to a biostate with dissolution like that, but

 

      I think those studies were done under different

 

      conditions.  These were done here, so they wouldn't

 

      have seen that under normal dissolution conditions.

 

                My more general question is--

 

                DR. YU:  You are correct, yes, in normal

 

      conditions, especially, for example, USP

 

      dissolution, maybe you are not able to see.

 

      Actually, dissolutions are beautiful.

 

                DR. MORRIS:  Right, so that comes back to

 

      sort of our discussions yesterday in a sense.  The

 

      question I have is to what level or to what extent

 

      do the ICH initiatives, I mean including the CTD

 

      and Q8, impact on the ANDA, I mean is there an

 

      intent that they follow suit with NDAs?

 

                DR. YU:  Obviously, the basic principles

 

      from ICH and CTD, the CTD cure document for drug

 

      substance and drug products, it is not just for NDA

 

                                                               111

 

      only, for both NDAs and ANDAs.  ICH Q8 is, in

 

      principle, a part into NDAs, certainly the basic

 

      principle also apply to ANDAs.

 

                The way I actively look into this to

 

      document and to see what information will help us

 

      to make scientific decisions.  Certainly, as I said

 

      before, we are not looking for information which is

 

      nice to know, we are looking for information which

 

      is essential to know.

 

                DR. MORRIS:  I guess to that end, because

 

      this is something, of course, we have been

 

      discussing for several years, but the idea that

 

      rather than having checklists of what the companies

 

      have to do, if they can make scientific decisions

 

      based on the intended dosage forms and the

 

      properties of the API, which should be a lower

 

      hurdle, I mean that should be known more by the

 

      time you get to the generic.

 

                DR. YU:  Yes.

 

                DR. MORRIS:  Instead of having to do a lot

 

      of the other testing that might normally be done,

 

      if they can focus on the identification of the

 

      critical to quality attributes of the product and

 

      capture that in a development report, it seems like

 

      that is a reasonable way forward.

 

                                                               112

 

                DR. YU:  That is correct.  In fact,

 

      industry is coming forward and they share some of

 

      the pharmaceutical development report with us, we

 

      are actively looking into this to develop some kind

 

      of review templates which will incorporate

 

      pharmaceutical development information into our

 

      review process.

 

                Again, I said we are looking for

 

      information which is essential to know, not nice to

 

      know.

 

                DR. MORRIS:  Maybe this is for Paul, is

 

      that a reasonable stance as far as how you look at

 

      generic development?

 

                DR. FACKLER:  I am not sure exactly what

 

      you are asking.

 

                DR. MORRIS:  I can clarify if you want,

 

                                                               113

 

      but basically, if you could, instead of having to

 

      do sort of checkbox testing, if you could do

 

      testing that was largely prescribed by your need to

 

      establish certain scientific issues, rather than

 

      having to do as many, let's say, sort of--what is

 

      the word--statutory testing, if you will, is that a

 

      reasonable stance for you guys?

 

                DR. FACKLER:  I don't see a problem with

 

      that.  What I didn't hear here was that there are

 

      any different statutory requirements for highly

 

      variable drugs.

 

                If a generic company still needs to pass a

 

      bioequivalence study, and we are going to assume

 

      that the pharmaceutical equivalence is simple, I

 

      don't understand what the generic company

 

      understanding the origin of the innovator's

 

      variability has to do with the approvability of a

 

      lot of material that is shown to be

 

      pharmaceutically equivalent and therapeutically

 

      equivalent through a bioequivalence study.

 

                I guess that is the piece I am missing.

 

      Why is the burden on the generic company to

 

                                                               114

 

      understand the variability of the reference listed

 

      drug, and what value does that have if really all

 

      the generic company needs to do still is

 

      demonstrate a bioequivalent product?

 

                DR. MORRIS:  You are talking about BE

 

      variability now, not pharmaceutical?

 

                DR. FACKLER:  Yes.

 

                DR. YU:  Paul, if we use

 

      one-size-fits-all, which is 80 to 125 percent to

 

      some of drugs, you may have difficulty to pass the

 

      confidence interval.  So, when we are exploring the

 

      alternative approach including the scaling

 

      approach, you will have to demonstrate this product

 

      is highly variable or not highly variable.

 

                You have to know that because otherwise,

 

      suppose someday in the future, if the scientific is

 

      mature enough, we have a scaling approach, for

 

      example, for highly variable drugs, your submitted

 

      application did not show these are highly variable

 

      drugs, how would we know these are highly variable

 

      drugs.

 

                So, you have to show, in your development

 

                                                               115

 

      report, that is a highly variable drug before we

 

      move forward.

 

                DR. FACKLER:  Agreed, but wouldn't a

 

      replicate design bioequivalence study inherently

 

      capture the variability of the reference listed

 

      drug?

 

                DR. YU:  Yes, if you choose to do so, use

 

      replicate design, certainly, you are able to

 

      demonstrate that reference list product is highly

 

      variable or not.

 

                DR. FACKLER:  But that is already part of

 

      an ANDA application is my point.

 

                DR. YU:  I guess, Paul, we have not

 

      reached a consensus or we have not made a

 

      determination you have to use replicate design.

 

                DR. MORRIS:  Is part of that the fact that

 

      you are still struggling with the concepts that are

 

      entailed in that dissolution plot where you can't

 

      factor into the pharmaceutical variability, factor

 

      the pharmaceutical variability from the clinical?

 

                DR. YU:  I guess the struggle we have here

 

      is, look, Lawrence, in order for you to get this

 

                                                               116

 

      direct for pass, whether you use scaling approach

 

      or you use widen the confidence interval, you

 

      simply widen the confidence interval, let them to

 

      pass.  You need to explain why.  You need to

 

      explain why you think that is a feasible approach,

 

      you think that is scientifically sound.

 

                So, when you say explain why the

 

      pharmaceutical development report can help us

 

      provide additional information to explain why.

 

                DR. FACKLER:  I agree that certainly you

 

      need to understand the variability of the reference

 

      listed drug especially if a generic applicant is

 

      claiming that the variability is an issue for this

 

      particular product.

 

                DR. YU:  Correct.

 

                DR. FACKLER:  I am not sure what value the

 

      steps that were taken has to that determination of

 

      variability.  Variability sometimes is listed in

 

      the label for a reference listed drug; other times

 

      applicants do replicate design studies or run

 

      reference versus reference to measure that inherent

 

      variability, but that would all be part of an

 

                                                               117

 

      application already, as I understand it.

 

                DR. YU:  Yes, in many cases actually

 

      lately for some of complex dosage forms.  Dosage

 

      form, we very often sent many, many deficiency

 

      letters.  Actually, company provide information

 

      during the cycles, and as I said, at the GPA Chair

 

      meeting, we have four or five or six cycles,

 

      provide additional information to us, and

 

      eventually, the product get approval.  I am not

 

      saying you not provide that information.

 

                What I am trying to say is with the arena

 

      of pharmaceutical development report in the ICHQ

 

      paradigm, can you provide that information in the

 

      application instead of for us to send many

 

      deficiency letters.

 

                When we see the OGD list receive 25

 

      percent or more of the applications every year,

 

      where do you want to put resource into those

 

      reviews.  Suppose you provide those additional

 

      information, which I believe will help us in our

 

      reviews, and they reduce the cycles, I see it's a

 

      win/win situation for you and for us.

 

                DR. COONEY:  Ajaz.

 

                DR. HUSSAIN:  I think look at it from this

 

      perspective in the sense the whole aspect is you

 

                                                               118

 

      are trying to make a decision and you are trying to

 

      choose the right measurement system here.

 

                Now, the Code of Federal Regulations

 

      essentially has a hierarchy of methods that you

 

      choose for bioequivalence.  Our current criteria is

 

      a PK crossover, PK-based, pharmacokinetic-based

 

      study is the most discriminating one.

 

                So, you are looking at, you are trying to

 

      now judge approvability of a generic drug, and for

 

      that you need to establish its pharmaceutical

 

      equivalence and its bioequivalence.  The

 

      bioequivalence measurement system that we have has

 

      inherent variability, and much of that variability

 

      is coming from the measurement system, and may not

 

      be coming from the test samples that you are doing.

 

                So, is this measurement system the ideal

 

      measurement system right now or not?  That is

 

      really the question.

 

                The dilemma that we have is the in vitro

 

                                                               119

 

      characterization and in vitro testing with

 

      dissolution often is not reliable enough by itself

 

      to make that call.  If it was, you would not be in

 

      this dilemma.

 

                So, if you really then look at it, what

 

      are we saying, is we have information generally

 

      that even if I give this drug intravenously or as a

 

      solution, and so forth, the variability is coming

 

      from the subjects, it is coming from physiology,

 

      which is inherently variable.  If I sleep on my

 

      lefthand side or righthand side, it will make a

 

      difference, I mean it literally happens.

 

                So, that is the measurement system, but

 

      then you are putting your product into that system

 

      and trying to see is there a difference of 20

 

      percent or not, and to meet that confidence

 

      interval criteria, you need 600 subjects or 300

 

      subjects, and so forth.

 

                Can we utilize the signs of design to say,

 

      to confirm, not necessarily to have a confidence

 

      interval, a confirmation that the new formulation

 

      actually is not contributing to that variability,

 

                                                               120

 

      is there sufficient science to do that or not.

 

                If it is, then it opens the door for

 

      saying that the bioequivalence assessment then

 

      could be tailored based on that understanding.

 

                DR. COONEY:  Before we go on to some other

 

      questions, I would like to see if your question,

 

      Paul, has been addressed.

 

                I think the question was--well, first,

 

      Lawrence is proposing that there be a

 

      pharmaceutical development report added to the

 

      information that is part of the application, and

 

      you are asking what will be the implications of

 

      providing that additional information and

 

      facilitating the next step, which is approval of a

 

      bioequivalence.

 

                DR. FACKLER:  That is part of the

 

      question.  The other part was what would be in a

 

      pharmaceutical development report that isn't

 

      already part of an ANDA.  That is really what I am

 

      trying to understand, and, of course, then, what

 

      value would that provide.

 

                DR. COONEY:  Is there clarity to that

 

                                                               121

 

      question?  So, that is back to Lawrence, to Paul's

 

      question.  What would be in that pharmaceutical

 

      development report that is not already part of the

 

      application?

 

                DR. YU:  I thought that was a topic of our

 

      next advisory committee meeting.

 

                DR. HUSSAIN:  Let me put it this way.

 

      There is nothing there right now.  There is nothing

 

      there to even gauge the aspects of.  So, what we do

 

      is our decisions are made based on one batch test

 

      results and the biostudy.  That is what it is.

 

                DR. MORRIS:  Can I just weigh in?  I think

 

      part of this is that a lot of what would go in the

 

      development report is stuff that people are already

 

      doing, but doesn't just get included in a summary

 

      fashion, much like we have discussed earlier, that

 

      there is development studies you do, but you don't

 

      put together.

 

                That is what we were talking about

 

      yesterday, is that, as a reviewer, if you have to

 

      try to piece together a development rationale from

 

      data here and there, you end up with sort of a

 

                                                               122

 

      development rationale that the person filing really

 

      wouldn't want to be there displayed to the world,

 

      you know, sort of a Frankenstein development

 

      report.

 

                So, if the company does it, then, they can

 

      see the logic that you use.  Whether they agree or

 

      not is a different question.  So, in my sort of

 

      concept of this, which may be flawed, of course, if

 

      the company, let's say, had used Cynthia's

 

      dissolution method, because they said this is what

 

      has really mattered, and they got those curves to

 

      overlay, then, that is a big step forward to say

 

      that the variability that may come out of the BE

 

      studies are not due to our change.

 

                So, if you see the variability of the BE

 

      studies and you have demonstrated that it is not

 

      due to the lack of adherence to a design space, for

 

      lack of a better word, then, that has got to be as

 

      good as the innovator.  That is my concept.  This

 

      may be down the road, as Jerry said.

 

                DR. YU:  I want to make comments that when

 

      we say the pharmaceutical development information,

 

                                                               123

 

      I think I emphasize those information that is

 

      essential to know, not just for nice to know.

 

                We are looking into this, what additional

 

      information will help us in making decisions, and I

 

      think we are happy to share with you in the future,

 

      but at this point, we cannot say that for every

 

      single ANDA or for every single product, you need

 

      the pharmaceutical developed, because you have a

 

      prior knowledge, some of the information already

 

      there, so this need clarification when you are

 

      understanding what additional information is

 

      provided.  I think we need to discuss and work it

 

      out.

 

                DR. FACKLER:  I understand.  To Ken's

 

      point, you start over here and the bioequivalent

 

      product is over here, and sometimes you take a

 

      direct approach to it and sometimes you don't.  You

 

      are right, oftentimes it is over here and then you

 

      realize you need to be over here, and then finally,

 

      you get where you need to be.

 

                But I am not sure I understand the value

 

      or what it matters what path you took as long as

 

                                                               124

 

      you end up in the right place.  All this

 

      information does exist, of course, and the field

 

      inspectors have access to it, and we are just

 

      reluctant to expand the content of an ANDA in the

 

      fear that it will slow down an already overburdened

 

      review process.

 

                So, where the information is critical to

 

      understanding whether a product is pharmaceutically

 

      or therapeutically equivalent, of course, it ought

 

      to be submitted, but where it is not essential for

 

      that evaluation, I just question whether or not it

 

      ought to be added to the burden of the reviewers.

 

                DR. COONEY:  Art, then Marvin.

 

                DR. KIBBE:  Let's get back to what we are

 

      trying to determine, and that is whether or not a

 

      clinician who prescribes this medication for its

 

      effect has got a reasonable expectation of a

 

      therapeutically similar outcome when he uses the

 

      innovator or when he uses the generic.  That is

 

      where we are.

 

                If a product is inherently variable, as

 

      manufactured by the innovator, then, we ought to

 

                                                               125

 

      know that early on, and as Les correctly points

 

      out, if that was the case during development and

 

      prior to approval, it wouldn't make it on the

 

      market if it wasn't that that breath of variability

 

      was allowable for clinical outcome, because if the

 

      clinical outcomes wouldn't--there were times when

 

      there were failures and times when they were

 

      toxicities apart, never gets on the market. which

 

      means that we have already historically established

 

      large variability is okay, because we have that

 

      product on the market.

 

                Now, if I am a generic company, all I want

 

      to do is say that I am going to be no more than, or

 

      perhaps less variable, and I am going to get to the

 

      same therapeutic outcome.

 

                If I can test a replicate design that

 

      shows that my level of variability is lower than or

 

      equal to the variability of the innovator, and my

 

      means are on target, then, I can with reasonable

 

      assurance argue that my product used in the

 

      marketplace on patients is going to have the same

 

      efficacy and failure rate as the innovator.

 

                The second thing is we already have agreed

 

      that dissolution is a hammer when we need a

 

      surgical scalpel to figure out what is going on,

 

                                                               126

 

      and if you make a shift in a dissolution criteria

 

      and all of a sudden you can differentiate tablets

 

      from the same batch, but that batch used in people

 

      isn't differentiatable, then, you are making a

 

      differentiation which is of no value to anyone

 

      except if you want to go back and process improve.

 

                In fact, that is what it should be used

 

      for.  The companies ought to be investing time and

 

      energy in process improvement by looking for better

 

      differentiators for their own internal consistency,

 

      and perhaps they could narrow the variability if

 

      they found them.

 

                I think the justification for going to the

 

      study that you said that if they used the USP

 

      numbers, they would all pass, and going to your

 

      numbers, we have this high variability, but that

 

      high variability doesn't relate to clinical

 

      outcomes.

 

                Now, I am coming on the market as a

 

                                                               127

 

      generic.  If I can establish that I am not more

 

      variable than they are, and my means are the same

 

      as they are in a biostudy, how much more

 

      information does the agency need?  I don't think it

 

      needs much more.

 

                DR. HUSSAIN:  Art, you are missing the

 

      point in the sense to demonstrate that your

 

      variability is acceptable, you actually have to do

 

      more now through a bioequivalence or replicate

 

      design, and so forth.

 

                What we are saying is in the sense, there

 

      are ways or there should be ways to sort of the

 

      justification that goes into a formulation that you

 

      move forward, could then become a basis to say you

 

      don't have to go through extraordinary means to say

 

      the variability is unacceptable.

 

                So, if we know a drug substance is highly

 

      variable, you mostly have that information that

 

      says you sort of at least definitely will when you

 

      approve the product, then, the signs of formulation

 

      design could provide you a basis for saying there

 

      is no reason your particle sizes, which are

 

                                                               128

 

      critical for your dissolution, your coating

 

      thickness, which are your release mechanism, are

 

      essentially being controlled, and so forth.

 

                So, why should a generic form then have to

 

      do a large study with replicate or with whatever?

 

      Isn't there an option available for something--

 

                DR. KIBBE:  So, what you are really

 

      talking about is a waiver of what we would say

 

      would be a standard replicate design to get around

 

      variability.

 

                DR. HUSSAIN:  Exactly, so that is what we

 

      are suggesting.

 

                DR. KIBBE:  So, the company then would

 

      come with its own development data and show that a

 

      broad range of dissolution numbers are not highly

 

      variable or something.

 

                DR. HUSSAIN:  Yes, the way I would think

 

      about that is in a sense if it's a tablet, I will

 

      go to the basic mechanisms of what the dissolution

 

      will be affected, and here is my assessment of my

 

      particle size, here is my control strategy, here is

 

      the prior knowledge of similar dosage forms.  There

 

                                                               129

 

      is no apparent reason for this to be variable from

 

      that perspective.

 

                So, that becomes a basis for a decision

 

      criteria saying that why would we expose normal

 

      healthy subject volunteers, a large number of them,

 

      to simply get our numbers within the confidence

 

      interval criteria, which is somewhat arbitrary.

 

      That is the crux of this.

 

                DR. YU:  I don't know if I can clarify,

 

      the point we are trying to make is that if you can

 

      conduct bioequivalence study now to best pass the

 

      confidence interval, this is good enough.  I am not

 

      saying this is not good enough.  We are not asking

 

      additional information.

 

                The problem which we are facing is you

 

      will not have difficulty, it is not impossible if

 

      you have recruited 1,000 or 2,000 subjects, it is

 

      almost impossible to do by a current study, and

 

      this is scenario that pharmaceutical variability

 

      information may come into play and to help us out.

 

      That is what we are trying to convey.  Thank you.

 

                DR. MEYER:  I think part of my problem is

 

                                                               130

 

      that I believe what you are putting forth is a

 

      concept without any data, which obviously you can't

 

      have yet, because the concept hasn't even been

 

      implemented, it is just a concept.

 

                I think certainly from my perspective, if

 

      you have some ideas that might streamline the whole

 

      system, I would say go for it and then let's see

 

      the meat once the skeleton is exhumed, so to speak.

 

                That is the bottom line, but I think there

 

      are some other ideas in there that are perhaps

 

      easier for me to understand, characterize the

 

      reference listed drug and then presumably, if you

 

      have done that, FDA will take that into

 

      consideration to explain why you have confidence

 

      limits that aren't up to par perhaps.

 

                For example, a simple example, the RLD has

 

      an overage in it of 10 percent.  They claim that

 

      isn't released ever, so they just have it in there

 

      because their release mechanism doesn't allow for

 

      except 100 percent.

 

                You have some evidence that says well, in

 

      fact, it is released 110 percent sometimes, so the

 

                                                               131

 

      poor generic company is already 10 percent in the

 

      hole when it comes to AUC.  If that can be

 

      demonstrated in some reasonable scientific fashion,

 

      that ought to maybe taken into account.

 

                A better example maybe is with the osmotic

 

      pump.  We have done studies where you can harvest

 

      the ghost out of the feces, and sometimes it has 50

 

      percent drug in it, sometimes it has 10 percent,

 

      sometimes it has no drug in it. It seems to be a

 

      direct function of intestinal transit time.

 

                Well, if you are a generic trying to match

 

      without using an osmotic pump, you don't have a

 

      snowball's chance in hell of coming across and

 

      matching a product that sometimes is 50, sometimes

 

      is 100, sometimes is 10 percent.

 

                So, I think as long as you hit the means,

 

      and you bring that kind of data to FDA, they ought

 

      to have the latitude of saying yeah, we know that's

 

      a problem with the RLD, and we can therefore adjust

 

      our thinking when it comes to the generic.

 

                Obviously, that is going to take a fair

 

      amount of work, but I think that these things need

 

                                                               132

 

      to be thought of, as well as more statistically

 

      based ways of dealing with high variability.  That

 

      is kind of a short-term fix which ultimately once

 

      the statisticians get done fighting, then, the rest

 

      of us can agree, but the other is certainly a

 

      concept worth pursuing, I think.

 

                DR. SINGPURWALLA:  I would like to respond

 

      to that.

 

                DR. HUSSAIN:  If I may, there is an aspect

 

      what Marv just said in the sense a practice that

 

      all of us know exists is when you have variability,

 

      then, you pick and choose what your comparator is.

 

      I mean it bothers me in a sense to say that, you

 

      know, you can pick and choose what lot you will

 

      compare to, and so forth.

 

                Why do we have to sort of have those type

 

      of decisions where, you know, I think we can be

 

      better than that, so I think just to build on what

 

      Marv says, to say that I think we can really be

 

      confident in what we are doing, and not to feel a

 

      bit guilty that we are picking and choosing what we

 

      test, and so forth.

 

                DR. MEYER:  As you well know I am sure,

 

      there are a number of countries.  You do your

 

      dissolution on three lots of the RLD and then you

 

                                                               133

 

      pick the one in the middle, not the one that is

 

      closest to what your product happens to be.

 

                DR. YU:  I want to make comments about

 

      Marvin's comments.  Yes, in the case here, what you

 

      present, actually, those information is not in the

 

      original ANDA submission, but those information

 

      eventually is shared with us.

 

                So, go through many cycles, many, many

 

      months, or even several years to get us that

 

      information.  What we are seeing is that we think

 

      if those information, which you eventually shared,

 

      only a couple that go through the five or six or

 

      seven cycles, shared in first place will help us to

 

      make decisions, will help us to reduce cycles, will

 

      help us actually use the resource wisely.  That is

 

      what we are trying to say.  Thank you.

 

                DR. COONEY:  Nozer.

 

                DR. SINGPURWALLA:  General comments.

 

      First thing, Ajaz, don't use the word confidence

 

                                                               134

 

      limits for those two boundaries.  Call them control

 

      limits.  Confidence limits are completely

 

      different.

 

                The second thing is you are fighting, at

 

      least there is a lot of discussion because there is

 

      a lot of variability.  What you seem to have done

 

      is taken reactive approach, have said variability

 

      is there, what shall we do about it.

 

                Well, yesterday, you talked about 6 sigma

 

      in one of your slides.  Well, I think wherever you

 

      have these high variability issues, whether they be

 

      in industry or whether they be within your own

 

      system, I would encourage you to put into practice

 

      what you were preaching yesterday about 6 sigma.

 

                I would say, you know, has anybody thought

 

      about that, because 6 sigma came about in industry

 

      because there was a lot of variability, and they

 

      said how do we control it.  Well, you just don't

 

      control it by doing statistical methods.  You

 

      control it by proper management and proper

 

      procedures, and I would say that you should try to

 

      bring that into the arena.

 

                DR. COONEY:  Paul.

 

                DR. FACKLER:  The generic industry is just

 

      as interested in minimizing the number of 6 and 7

 

                                                               135

 

      cycle reviews on products.  Clearly, we have the

 

      same goal in mind.

 

                I guess what I would suggest is that for

 

      highly variable drugs, for instance, it would be

 

      useful for the agency to tell industry the kind of

 

      information that is generally lacking, but with 500

 

      applications a year, or 800, whatever the numbers

 

      might be these days, coming into the agency, I

 

      don't think it is wise to require this information

 

      on all of the applications.

 

                I would suggest maybe we clarify the

 

      additional information that is often being left out

 

      of submissions for highly variable products, and

 

      presumably, generic companies in the interest of

 

      having a minimum number of review cycles will

 

      submit it the first time rather than an iterative

 

      process to give you all the information that you

 

      need to make a fair decision.

 

                DR. COONEY:  Gary.

 

                DR. BUEHLER:  For the development reports

 

      in general, I thank you for not wanting to

 

      overburden us with additional information.  We do

 

      have a lot to look at.  If we do get additional

 

      information, we will look at it for sure.

 

                I know that we get some amount of this

 

                                                               136

 

      information sprinkled through the ANDAs and I would

 

      think Ajaz was a bit draconian when he said all we

 

      get is the batch record and whatever.  I mean there

 

      are explanations.  We do demand explanations when

 

      there aren't any deviations from what we normally

 

      see, that is in ANDAs and we do look at that.

 

                Lawrence and a group is working on a

 

      question-based review for the Office of Generic

 

      Drugs.  It is a very detailed project.  He is

 

      working with experienced reviewers in our office,

 

      and he is developing this in a very stepwise

 

      manner, both first by involving both the

 

      supervisors and reviewers in our own office, and

 

      then at a certain point we want to sort of unveil

 

      it to industry.

 

                We want to make sure that when we do bring

 

                                                               137

 

      this new review method and these new requirements

 

      or whatever you want to call them with respect to

 

      pharmaceutical development reports, the industry is

 

      very aware of what we want and why we want it, so

 

      that they will feel good about giving us this

 

      information, and like Lawrence said, it will

 

      hopefully reduce the number of cycles we have, it

 

      will not overburden the reviewers, but, in fact,

 

      reduce the burden on the reviewers, because they

 

      won't have to see the same applications four, five,

 

      or six times, and they will understand why we need

 

      this information.

 

                It is also a risk-based system, so that

 

      there are some applications that you won't have to

 

      provide this type of information, because there are

 

      some applications obviously that are easier than

 

      other applications, and the applications for

 

      complex dosage forms and unique dosage forms

 

      obviously, we are going to ask for more information

 

      than for the vary standard solid orals that are

 

      fairly easy to manufacture.

 

                But we are doing this over a two-year

 

                                                               138

 

      period and hopefully, sometime toward the end of

 

      this year, we will be able to begin to tell

 

      industry what we hope to expect in the future

 

      applications and industry will be comfortable with

 

      it.

 

                DR. MORRIS:  I just have a quick question

 

      for Gary.  I am assuming that development reports,

 

      as you say, depending upon the complexity of the

 

      dosage form, I mean they can be relatively brief if

 

      it's a very simplistic or simple dosage form, so I

 

      am not so sure that it's the burden if the payback

 

      is fewer review cycles or less clinical studies.

 

      Clinical studies are a lot more expensive than

 

      writing a development report and doing a few more

 

      development studies.

 

                Is that more or less the case, Gary?

 

                DR. BUEHLER:  I am not sure it is going to

 

      be able to be submitted in lieu of a study or

 

      whatever.

 

                DR. MORRIS:  No, I meant the extensiveness

 

      of a development report.

 

                DR. BUEHLER:  Some development reports

 

                                                               139

 

      will say we wanted to develop a bioequivalent

 

      formulation, and, you know, here it is, and it

 

      could be a page or two.  I mean clearly, it won't

 

      be very long for a generic, because the goal of a

 

      generic is pretty evident, but other development

 

      reports will be more extensive, so yeah, you know.

 

                DR. COONEY:  It sounds like there is a

 

      need for clarity on what will be requested and

 

      expected, and also for clarity on what the

 

      implications of that will be.  It sounds like that

 

      will be forthcoming.

 

                Ajax, what I would like to do is move on

 

      to the next presentation.

 

                DR. HUSSAIN:  Just go back to the original

 

      intent.  Our initial thoughts that we wanted to get

 

      the discussion started, so we never intended this

 

      to make a proposal, so these are initial thoughts

 

      and we are moving forward with this.

 

                If industry wants to be proactive, they

 

      had better start thinking about it and how they can

 

      use this opportunity instead of asking us what do

 

      we want.  I think it is equally burdensome on

 

                                                               140

 

      industry to think about how to develop the products

 

      for the intended use, and make the case, and grab

 

      that opportunity.

 

                If not, the system as it stays, we are

 

      perfectly happy with it.

 

                DR. COONEY:  So, there is an opportunity

 

      here for dialogue and there is no doubt from the

 

      last 45 minutes that there will be dialogue.

 

                I would like to ask Robert Lionberger to

 

      proceed with the next presentation.

 

            Using Product Development Information to Support

 

                Establishing Therapeutic Equivalence of

 

                            Topical Products

 

                DR. LIONBERGER:  Today, I am going to be

 

      discussing how to apply the concepts of

 

      pharmaceutical equivalence to topical dosage forms

 

      and look at how this is related to quality by

 

      design.

 

                Here, I am going to focus on topical

 

      dosage forms that are in the local delivery, so not

 

      products such as transdermal products that are

 

      trying to deliver drugs systemically.

 

                In the Office of Generic Drugs, as you

 

      have heard several times before this morning, our

 

      mission is to provide therapeutically equivalent

 

                                                               141

 

      products to the public.  When someone uses a

 

      generic drug, they should expect the same clinical

 

      effect and safety profile as the branded reference

 

      product.

 

                Just to summarize some things that Ajaz

 

      talked about in his introductions, the preface to

 

      the Orange Book explains how we do that.  Products

 

      must be pharmaceutically equivalent and

 

      bioequivalent.  But I want to dig a little bit

 

      deeper into this and ask why do we actually require

 

      both, why isn't bioequivalence by itself enough to

 

      determine that the products are the same.

 

                One aspect of that is that consumers have

 

      some expectation about product behavior.  If the

 

      reference product is a capsule, you don't want to

 

      replace that with a solution.  So, there is some

 

      user experience and expectation.

 

                So, pharmaceutical equivalence

 

      encapsulates concepts related to like the user

 

                                                               142

 

      interface of the product, but then there is another

 

      aspect to it, and I have tried to express it here,

 

      is that pharmaceutical equivalence supports the

 

      determination of therapeutic equivalence based on

 

      bioequivalence study.

 

                We don't say that just because two

 

      products pass a bioequivalence study, they are

 

      therapeutically equivalent products.  An example

 

      might be an oral solution and a tablet.  There can

 

      be many products for which those two dosage forms

 

      would be bioequivalent, but we wouldn't say that

 

      they are therapeutically equivalent products.

 

                One aspect of that is that our current

 

      determination of bioequivalence is really very

 

      strongly based on in vivo testing.  So, again,

 

      there are limitations to testing.  We test these

 

      products in a small population  and then we

 

      extrapolate that conclusion to all people who are

 

      going to use the products from all batches in the

 

      future.

 

                So, to sort of back up that extrapolation,

 

      there is some other information.  Right now that's

 

                                                               143

 

      the pharmaceutical equivalence between the products

 

      that supports that.

 

                In the occasions when we do equivalence

 

      studies in patients, there are other differences.

 

      Sometimes the clinical endpoints aren't very

 

      sensitive to small differences, bringing in

 

      examples from topical products, you can imagine

 

      there are cases where, say, a cream and an ointment

 

      formulation might have the same therapeutic effect,

 

      but they wouldn't be considered pharmaceutically

 

      equivalent products or therapeutically equivalent

 

      products even though the clinical endpoint study

 

      might show equivalent efficacy.

 

                Again, from the sort of pharmacokinetic

 

      studies for one of the challenges that is often

 

      made to some of our bioequivalent studies for

 

      topical products is since the skin is a barrier,

 

      you say, well, healthy subjects have healthy skin

 

      barriers.  There is a question.  Sometimes people

 

      will claim in patients, the skin might be diseased

 

      or damaged, so that is a common concern.  There is

 

      a common challenge to some of our bioequivalence

 

                                                               144

 

      determinations here.

 

                So, inside of the pharmaceutical

 

      equivalence concept, there is some idea of other

 

      things we want to know about the products to sort

 

      of generalize this idea of equivalence.

 

                If you think about this and want to sort

 

      of tie this to quality by design, one way that

 

      might be useful for you to think about this is that

 

      our current definition of pharmaceutical

 

      equivalence might be considered a first step toward

 

      a quality by design.

 

                If you were going to design equivalent

 

      products, the first things you would start with

 

      were some of the concepts that are in our current

 

      definition of pharmaceutical equivalence.  You

 

      would want to have the same active ingredient.  You

 

      would want to have the same strength, the same

 

      dosage form.

 

                So, if we look at sort of a different way

 

      of looking at our paradigm, maybe instead of a

 

      regulatory framework, a more scientific framework,

 

      what we are doing when we review a generic product,

 

                                                               145

 

      is we are looking to see is the product designed to

 

      be equivalent, and then does it demonstrate

 

      bioequivalence in an in vivo study.

 

                So, you can see sort of this combination

 

      sort of parallels our current sort of regulatory

 

      framework of pharmaceutical equivalence and

 

      bioequivalence leading to a determination of

 

      therapeutic equivalence, where we might say that on

 

      sort of a scientific level, what we might want to

 

      be doing in the future might be to say look at the

 

      quality by design, look at the generic product that

 

      is designed to be equivalent to a reference

 

      product, and then based on this design, choose the

 

      appropriate either in vitro or in vivo

 

      bioequivalence testing for this product to complete

 

      the determination of therapeutic equivalence.

 

                So, I want to bring this sort of

 

      conceptual framework and bring it into this sort of

 

      particular example for topical products.  Sort of

 

      to motivate that, I just want to outline some of

 

      the complex issues that we deal with that are

 

      related to pharmaceutical equivalence for topical

 

                                                               146

 

      products.

 

                Again, we have a lot more experience with

 

      immediate release, oral dosage forms in effective

 

      excipients, what excipients you can change, what

 

      excipients you can't change.  For topical products,

 

      a lot of times the excipients may or may not affect

 

      the barrier properties of the skin and drug

 

      delivery.

 

                We don't have as much experience about

 

      that, so a lot of times we are worried about what

 

      differences in formulation are appropriate for

 

      comparing a test in a reference product - is a

 

      change in solvent appropriate, what if the base of

 

      the formulation in ointment or cream has changed

 

      from being hydrophilic to lipophilic, how much

 

      water content should there be in the product.  You

 

      might affect evaporation, the feel of the product.

 

                A lot of these sort of differences in

 

      formulation get wrapped up into the question of are

 

      two products the same dosage form.  I will talk a

 

      little bit more in detail about that in the rest of

 

      the products.

 

                We also have questions, when we don't have

 

      good bioequivalence methods for use for topical

 

      products, what indications should be used for the

 

                                                               147

 

      clinical equivalence studies.  Perhaps the product

 

      has multiple indications, which one is the most

 

      appropriate one to use.

 

                These are the kinds of issues that we deal

 

      with in generic topical products.  Some of the

 

      implications of these for the ANDA sponsors are

 

      that the approval times for these products can be

 

      longer.  If there are these issues that we don't

 

      have a good understanding internally, we have to

 

      schedule meetings with the appropriate people, have

 

      to have internal discussions.

 

                When the sort of standards aren't clear,

 

      this is an opportunity for the reference listed

 

      drug sponsors to challenge correspondence to OGD or

 

      through the citizen petition process that we have

 

      to address the scientific issues there that aren't

 

      sort of clearly defined.

 

                A lot of times, at the end of these

 

      discussions, we will end up going back to the

 

                                                               148

 

      sponsors and asking them for more information to

 

      help us resolve these issues, and then usually that

 

      is done through deficiency letters to them, and it

 

      ends up with sort of multiple review cycles.

 

                So, as we heard in Lawrence's talk, there

 

      is the question of more product development

 

      information in the ANDA itself may help OGD deal

 

      with these issues more efficiently.

 

                This is sort of very similar to some of

 

      the things that Lawrence talked about, that there

 

      are harmonization efforts underway that describe a

 

      product development report, but I think it is clear

 

      that these are mainly aimed at new drug

 

      applications, so it is not sort of obvious or clear

 

      how these should apply to ANDA sponsors.

 

                I think the theme of this talk to see this

 

      as an opportunity, these development reports, as an

 

      opportunity to provide information that will help

 

      the agency set rational specifications for products

 

      that are complex, for immediate release oral dosage

 

      forms we have various standard systems set in

 

      place, but for topical products, where we have less

 

                                                               149

 

      experience, the more information that is provided

 

      about, say, why was an excipient changed, and why

 

      do you know that it is not going to have any effect

 

      could be very helpful to us in making efficient

 

      decisions.

 

                Again, the product development reports are

 

      the place in the application to emphasize the

 

      quality by design, that the product is designed to

 

      be equivalent.  That will help us set the right

 

      requirements for the bioequivalence testing for

 

      particular products.

 

                This is just a few examples of what some

 

      of these harmonization documents say about a

 

      pharmaceutical development report.

 

                In this case, again, the key part here

 

      might be to establish that the dosage form and the

 

      formulation are appropriate for the purpose

 

      specified in the application, or in the Q8

 

      document, it talks about an opportunity to present

 

      the knowledge gained through the application of

 

      scientific approaches.

 

                Here, it is talking specifically about

 

                                                               150

 

      sort of formulation and development for the topical

 

      products, that there is information that the

 

      company that is developing the generic product

 

      knows about why they made certain choices in the

 

      formulation.  It would be very helpful to us in

 

      deciding that that is acceptable, where the agency

 

      itself has less experience with particular dosage

 

      forms.

 

                I have emphasized this concept of quality

 

      by design or, in the case of the generic products,

 

      quality by design means you are designing the

 

      product to be equivalent to the reference product.

 

                So, I want to try to be a little bit more

 

      specific about what that means.  There are two

 

      cases.  One, the mechanism of release.  Clearly,

 

      the mechanism of release between a generic product

 

      and the reference product can be different, but the

 

      intent of those different mechanisms ought to be to

 

      produce the same rate and extent of absorption.

 

      This is the bioequivalence criteria.

 

                Again, we also recognize that depending on

 

      the particular product, that the release rate from

 

                                                               151

 

      the product might not be the rate controlling step

 

      at absorption.  So, the determination of how close

 

      release rates might have to match would depend on

 

      the absorption process involved and what is the

 

      rate-limiting step in the absorption process.

 

                Again, between generic products and

 

      reference products, the excipients can be

 

      different.  Again, it is a good thing to understand

 

      the differences between the excipients.

 

                The IIG limits are a starting point.  They

 

      tell you that this excipient has been used in this

 

      dosage form up to a certain amount, and that really

 

      addresses, specifically in the case of topical

 

      products, safety-related exposures, so you know

 

      that level of exposure.

 

                The thing that complicates the topical

 

      products is when you change the excipients, the

 

      real question that we often deal with is do the

 

      changes in the excipients to the products affect

 

      the permeation of the drug through the skin. I

 

      think that is the sort of challenging question

 

      there for the topical products that we occasionally

 

                                                               152

 

      have to deal with.

 

                Again, as I said, the purpose of quality

 

      by design is to design the equivalent product.  I

 

      want to just give sort of three sort of examples of

 

      this process here.

 

                The first is talking about Q1 and Q2

 

      equivalent products for topical products, and then

 

      look at what happens when you make changes to the

 

      formulation, they become Q1 and Q2 different, and

 

      then this leads into the discussion of issues

 

      related to the dosage form classification and how

 

      product development information might help us make

 

      a better decision or more scientific based

 

      decisions on dosage form classifications.

 

                First, I want to start off with the

 

      definition of Q1, Q2, Q3.  So, products that are Q1

 

      have the same components, so both the generic and

 

      the reference product would have the same

 

      components.

 

                If products are Q2, they would have the

 

      same components, but they would also have the same

 

      amount of each ingredient.

 

                The Q3 concept is same components, same

 

      concentration, but here I am saying same

 

      arrangement of material or microstructure, and this

 

                                                               153

 

      is particularly important for topical products that

 

      are semisolid dosage forms, so non-equilibrium

 

      dosage forms, where you might have, say, an

 

      emulsion with exactly the same components, exactly

 

      the same concentrations, but say, for example, the

 

      droplet size might depend on how you have

 

      manufactured that product.

 

                So, there is potential differences for

 

      semisolid dosage forms depending on how they are

 

      produced even if overall the composition is exactly

 

      the same.

 

                A contrary example would be a solution.

 

      If a solution is Q1 and Q2, because the solution is

 

      at thermodynamic equilibrium, you would be able to

 

      say we know that this product has exactly the same

 

      arrangement of material in the product.

 

                The importance of the Q3 concept is when

 

      you know that the products have the same

 

      arrangement of material, you know that they are

 

                                                               154

 

      going to be bioequivalent, there is no question

 

      about that.  An example of that is again a solution

 

      where you know that the products are in

 

      thermodynamic equilibrium if the compositions are

 

      the same, the structure and arrangement of the

 

      material is the same.

 

                Unfortunately, for most topical semisolid

 

      dosage forms, they are not necessarily equilibrium

 

      arrangements of material, and so a direct

 

      measurement of Q3 level equivalence is challenging.

 

                So, if we have the topical products where

 

      Q1 and Q2 are identical, again, the only potential

 

      differences are differences in this Q3 parameter,

 

      which can come from differences in manufacturing

 

      processes, because they are not going to be

 

      manufactured by exactly the same process.

 

                We know for particular semisolid dosage

 

      forms, such as emulsions, that rheology and in

 

      vitro release rates can be very sensitive

 

      measurements of microstructure and are related to

 

      product performances.

 

                So, the sort of idea that sort of

 

                                                               155

 

      advancing here from a scientific point of view is

 

      when the products are Q1 and Q2, that in vitro

 

      tests should be equivalent to ensure bioequivalence

 

      of the two products, because again here, the issues

 

      are detecting differences due to differences in

 

      manufacturing processes, and the argument would be

 

      that in vitro tests are the best evaluation method

 

      to detect whether any differences in manufacturing

 

      process have significant differences in the product

 

      formulation or performance.

 

                Now, things get more complex when a

 

      generic product and a reference product have

 

      different compositions, and this connects with the

 

      dosage form classification, and these differences

 

      occasionally could be barriers to generic

 

      competition.

 

                A generic company might want to formulate

 

      products that are Q1/Q2 different because the

 

      innovator has formulation patterns, so there might

 

      be either legal reasons or perhaps manufacturing

 

      process reasons why you might want to formulate a

 

      product that is not exactly identical in

 

                                                               156

 

      composition to the reference product.

 

                One of the products, again, one of the

 

      members of the Committee mentioned the sort of

 

      economic effect of uncertainty on product

 

      development if you don't know what dosage form the

 

      product is going to be classified as.  That adds

 

      cost to the development process because of

 

      uncertainty of what is going to happen to the

 

      product.

 

                In particular, if we think about methods

 

      by which we would classify the dosage form of

 

      topical products, here, I have generated a list of

 

      four possible ways that you could approach this.

 

                One is we would just use whatever the

 

      sponsor says their product is as long as it is

 

      consistent with some of the traditional definitions

 

      that are available in various sources, and we will

 

      look in sort of detail at some of those traditional

 

      definitions.

 

                You might say, well, the generic product

 

      is the same dosage form if it feels the same to me,

 

      so I will just try it out and see if it is the

 

                                                               157

 

      same, if it passes.  You know, if the look and feel

 

      of the products are the same. That is getting to

 

      the aspect of pharmaceutical equivalence, a sort of

 

      patient experience rather than sort of scientific

 

      issues related to product performance.

 

                Then, I am going to sort of discuss recent

 

      work that the FDA group led by Cindy has done on

 

      looking at a whole bunch of products and coming up

 

      with a quantitative decision tree to classify

 

      topical products.

 

                Then, sort of the fourth aspect of that is

 

      looking at whether or not issues about dosage form

 

      classification for complex issues would be

 

      something that you would want to include in a

 

      product development report, so justifying the

 

      formulation development as being the same as the

 

      reference product.  That sort of might be a more

 

      scientific way to look at these issues.

 

                First, if we look a some of the

 

      traditional definitions.  Here, I will just focus

 

      on the difference between a cream and an ointment.

 

      One source is the CDER's data standards

 

                                                               158

 

      definitions.  These are sort of similar to USP

 

      definitions of these products.

 

                The cream is a semisolid dosage form

 

      containing one or more drug substances dissolved in

 

      a suitable base, and then it says more recently the

 

      term has been restricted to products consisting of

 

      oil-in-water emulsions.  That is obvious what a

 

      company should do - does a cream have to be an

 

      oil-in-water emulsion or not.

 

                Then, it talks about products that are

 

      cosmetically and aesthetically acceptable, is part

 

      of the definition of the cream, so that is not very

 

      quantitative. It is hard to say is this product

 

      aesthetically acceptable. That is really opinion

 

      based.

 

                An ointment is a semisolid preparation

 

      intended for external application.  It seems to me

 

      that a cream could be a semisolid preparation and

 

      fit under the ointment definition.  So, it doesn't

 

      seem that those two definitions are really

 

      exclusive.

 

                In another FDA guidance, this is the SUPAC

 

                                                               159

 

      semisolid guidance.  It has a glossary with

 

      definitions of dosage forms, but these aren't the

 

      same as the previous ones.  A cream is a semisolid

 

      emulsion, and an ointment is an unctuous semisolid

 

      and typically based.  So, typically based is not

 

      sort of a definition, it doesn't have to be based

 

      on petrolatum.

 

                This definition talks about an ointment

 

      being one phase, and not having sufficient water.

 

      Again, the USP definition is sort of similar to the

 

      one in the CDER data standards, but it is not

 

      word-for-word identical, and talks about four

 

      different classes of ointments.

 

                So, again, the problem with the

 

      traditional classifications is they are not really

 

      consistent, and not very quantitative.  So, a lot

 

      of the sort of decision process would depend on

 

      what your opinion was of a particular product, and

 

      they might be overlapping, like you might be able

 

      to call--under a particular definition of a

 

      particular product, you might be able to call it a

 

      cream or an ointment.

 

                So, the result of this Topical Working

 

      Group led by Cindy has been presented to previous

 

      advisory committee meetings, and they recently

 

                                                               160

 

      published a paper outlining this classification

 

      scheme.

 

                What they did was they surveyed existing

 

      products and devised a classification scheme, and I

 

      just included the classification scheme here just

 

      for reference in the presentation.

 

                These are just some slides from their

 

      previous presentations to give you the general idea

 

      of what they did. They measured particular aspects

 

      of products, say, creams and ointments, they

 

      measured viscosity.

 

                They looked at the loss on drying, and

 

      then based on these products that were either on

 

      the market or manufactured for them, they came up

 

      with a classification scheme that sort of put the

 

      products in the right category based on existing

 

      products.

 

                The real advantage of this is it is

 

      quantitative. If you take a product and you go

 

                                                               161

 

      through and you measure the things outlined in

 

      their decision tree, you will always end up in the

 

      same place, and it will always be consistent.

 

                In addition to that, this is a very data

 

      driven approach.  They looked at the products and

 

      then drew the lines.  It wasn't said here is sort

 

      of a mechanistic definition of what a cream or an

 

      ointment should do.

 

                So, the question is, could this be overly

 

      restrictive.  If you follow this classification

 

      scheme, you would be restricting products to

 

      essentially what has been done before, and then

 

      there is a question.

 

                They didn't survey every product that is

 

      on the market now, so there is a question, if a

 

      reference listed drug falls into a different part

 

      of this classification scheme, then, it's labeling.

 

      So, it might be labeled as a cream, but by the

 

      definition, it would be classified as an ointment.

 

      What should a sponsor do in that case?

 

                So, the final sort of approach to dosage

 

      form classification might be to look at a more

 

                                                               162

 

      scientific view of the formulation design.  I just

 

      want to point out that sort of the legal aspects,

 

      referring to topical use, sort of point toward this

 

      here from the CFR.

 

                It talks about inactive ingredient changes

 

      for topical products.  It says again that

 

      abbreviated applications can use different

 

      ingredients if they identify and characterize the

 

      difference and provide information demonstrating

 

      that the differences do not affect the safety or

 

      efficacy of the proposed drug products.

 

                So, a current way of looking at is a

 

      change in formulation acceptable, you should check

 

      the new excipients against the IIG.  As I said

 

      before, this looks at the safety of the individual

 

      excipients.

 

                We also really consider that passing

 

      bioequivalence tests are evidence that the

 

      formulation change is acceptable.  That is one

 

      strong piece of evidence against that.  But again

 

      the product development report is an opportunity

 

      for sponsors to characterize the differences.

 

                Again, this could be important, you know,

 

      if you are formulating a product and you are on the

 

      boundaries of these, we have this empirical

 

                                                               163

 

      decision tree, what happens if you are on the

 

      boundaries, how do you explain that this product

 

      should be considered the same as the reference

 

      product, you know, from a scientific point of view

 

      rather than empirical classification scheme, of if

 

      someone says, well, no, your product is not really

 

      an ointment because it doesn't meet a particular

 

      published definition.

 

                So, again, the product development report

 

      is the opportunity for a sponsor to characterize

 

      the difference that is sort of requested in the

 

      statutes.

 

                Again, also, in the statutes, they list

 

      reasons to reject ANDAs, and they talk about drug

 

      products for topical administration where there is

 

      a change in lipophilic properties of the vehicle.

 

                Again, in this case, a product development

 

      report is an opportunity for sponsors to explain

 

      why the changes, Q1 and Q2 differences are

 

                                                               164

 

      appropriate for this particular product.

 

                If this issue comes up, a lot of times we

 

      will actually have to go from the review process,

 

      you will have to go back and ask sponsors for more

 

      information about these particular issues.  So, the

 

      development report is sort of an up-front way to

 

      explain the reasons for doing that.

 

                Just to sort of conclude the discussion

 

      here, the first concept is the importance of Q1,

 

      Q2, Q3 classification to identify appropriate

 

      bioequivalent studies for the level of difference

 

      in the product design.  If a few products have

 

      exactly the same active and inactive ingredients,

 

      you might want to request different in vivo

 

      bioequivalence studies than for a product where

 

      there has been a change in inactive ingredient that

 

      may affect the absorption of the drug product.

 

                So, here again, we are looking at the

 

      second concept, we are looking at the evolution of

 

      the concept of pharmaceutical equivalence where we

 

      have these traditional dosage form definitions,

 

      maybe now backed up by empirical decision trees,

 

                                                               165

 

      but in the future, looking for a quality-by-design

 

      aspect where the determination of whether a product

 

      should be considered equivalent would depend on the

 

      mechanistic understanding and the formulation

 

      design rather than some traditional definitions,

 

      and that the ideal state would be that this

 

      understanding would reduce the need or allow us to

 

      set the appropriate in vitro testing for a

 

      particular product, and also to expand sort of the

 

      formulation design space beyond past experience.

 

                If you want to formulate a product that

 

      goes beyond, say, an empirical dosage form

 

      classification, this is the sort of way that you

 

      would approach it, by providing the scientific

 

      information to show that the formulation you have

 

      chosen gives equivalent performance in the key

 

      attributes as the reference product.

 

                With that, I will conclude my

 

      presentation.

 

                DR. COONEY:  Thank you.  I believe the

 

      purpose of your presentation today is to bring us

 

      up to date on the current thinking where you are

 

                                                               166

 

      and where you are going as opposed to requesting a

 

      specific action on our part, is that correct?

 

                DR. LIONBERGER:  Yes, that's right.

 

                DR. COONEY:  I would like to invite

 

      questions and comments from the Committee.  Yes,

 

      Cynthia.

 

                DR. SELASSIE:  This is a very general

 

      question.  With all these product development

 

      reports that you get, obviously, there is going to

 

      be a lot of information that is extraneous and

 

      won't be useful for that particular application,

 

      but will you all retain this information like in a

 

      database, so that it could have use down the line?

 

                DR. LIONBERGER:  I don't know if we would

 

      retain it in a database, but I would say that like

 

      as Lawrence said, we are looking at our review

 

      process, and in that, had the opportunity to read

 

      several product development reports.

 

                I find that they are a very useful way to

 

      get an overview of what is going on with a

 

      particular product.  You know, an hour of reading

 

      the development report, it seems like a very good

 

                                                               167

 

      way to start the review of the application in more

 

      detail, so I think it can be very valuable.

 

                We don't have much experience with using

 

      them yet, so in that sense, it could be valuable,

 

      but we don't know how we would use that information

 

      or store that information in the future.

 

                DR. HUSSAIN:  If you are suggesting that

 

      there is a need to capture and create databases, I

 

      think we do want to move in that direction, and we

 

      tried to do that.  Currently, our systems does

 

      capture some of the key aspects. The inactive

 

      ingredient guide is a process that we capture every

 

      inactive ingredient that comes, but developing a

 

      formal knowledge base would really be helpful, and

 

      I think we have been thinking about it.

 

                I tried to do that with immediate release

 

      dosage forms and actually did some modeling with

 

      that data that we have, and so forth, so we will

 

      look into that.

 

                DR. COONEY:  Are there any other questions

 

      from the Committee?  Marv.

 

                DR. MEYER:  Just a quick comment.  You

 

                                                               168

 

      have my sympathies.  I thought it was difficult to

 

      determine how to do the BE studies on topicals, and

 

      now I have been reinforced, you don't even know

 

      what slot to put them in the Orange Book if they

 

      are bioequivalent, so you have a big job ahead of

 

      you.

 

                DR. COONEY:  If there are no further

 

      comments, Robert, thank you very much.

 

                There have been no requests for

 

      participation in the open public hearing at 1

 

      o'clock, so we will proceed with the continuation

 

      of the discussion on quality by design precisely at

 

      1 o'clock when we come back from lunch.

 

                We will begin that by a presentation of a

 

      summary of the plan by Ajaz, and then we will

 

      continue the discussion that we began earlier this

 

      morning.

 

                So, enjoy lunch and we will see you back

 

      at 1 o'clock.

 

                [Whereupon, at 11:59 a.m., the proceedings

 

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

 

                                                               169

 

                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. COONEY:  I would like to welcome

 

      everyone back from lunch.

 

                We will proceed with the afternoon

 

      schedule.  The first topic this afternoon will be

 

      Ajaz Hussain, who will provide a summary

 

      description of the plan to go forward.

 

                  Quality-by-Design and Pharmaceutical

 

                        Equivalence (Continued)

 

                            Summary of Plan

 

                DR. HUSSAIN:  I am going to go back to the

 

      slides I used in the morning instead of the ones I

 

      had for this session.  That was based on the

 

      discussion that occurred.

 

                Just on reflection, I just want to make a

 

      couple of points.  Yesterday, in a sense, as part

 

      of the tactical plan to start our journey towards

 

      the desired state, in a sense what we have done at

 

      this meeting is to take a look back last 10 years

 

      or so to see how our policies have evolved and how

 

      they could evolve with two tools that we have

 

                                                               170

 

      introduced, the PAT guidance and ICH Q8.

 

                Tom Layloff reminded me that in many ways,

 

      some of the topics we have discussed, we have been

 

      discussing for 30 years, and we keep discussing

 

      those topics again and again, and the difference

 

      that we have tried, at the training session that

 

      some of you attended, I am clearly cognizant of the

 

      fact that we are discussing topics that we have

 

      been discussing for 30 years, and the quote I had

 

      was the thing that if you tried to approach the

 

      problem with the same tools and the same approach

 

      again, we are bound to find the same solutions, so

 

      we need something new.

 

                What is new at the issue of this problem

 

      is the science of formulation design, of science of

 

      product design. The key aspect, much of that has

 

      always been considered as an art, and as the

 

      complexity of products is increasing, that art will

 

      not be sufficient to really achieve the performance

 

      we are trying to achieve.

 

                So, it is a reflection back of saying all

 

      right, 30 years of pharmaceutical sciences in

 

                                                               171

 

      particular pharmaceutics, industry, pharmacy, and

 

      so forth, what have we learned and what we need to

 

      learn more to do things differently.

 

                In some sense, that is the heart of the

 

      debate.  I also sort of mentioned to you, and this

 

      is my original starting point in the thought

 

      process was that you really need at FDA more people

 

      with that background to really make that happen.  I

 

      changed my thoughts over the last several years.

 

                What we have at FDA is scientists from

 

      many, many different disciplines who sort of work

 

      together.  The reason I changed my mind was I think

 

      looking at some of the practices and formulation

 

      development, and so forth, you really need a

 

      multidisciplinary approach to challenge some of the

 

      inherent assumptions which are in the system.

 

                Therefore, I think what we have is

 

      non-pharmaceutics people evaluating this is an

 

      advantage, not a disadvantage, but then the key is

 

      you have to put this in a scientific terminology

 

      that can become negated across different scientific

 

      disciplines.  That is a significant challenge.

 

                So, with that in mind, we want to make

 

      this a scientific process.  The review assessment

 

      is a scientific process.  Therefore, it has to be

 

                                                               172

 

      essentially a scientific format offer hypothesis

 

      tested.

 

                So, with that in mind, if I look back at

 

      the SUPAC guideline, what we have done there is the

 

      guideline was a first step in moving towards this

 

      direction, and in a sense we tried to identify in

 

      that guideline changes that can be classified as

 

      minor, moderate, and major changes.

 

                How did we accomplish that?  We

 

      accomplished that through expert solicitation is a

 

      very real thing, not just where we had some

 

      workshops, and where we collected the wisdom of

 

      people in this area to say what are the changes

 

      which are minor, major, and so forth.

 

                Then, we took those recommendations and

 

      actually challenged those recommendations through

 

      experiments and studies that we did at University

 

      of Maryland to design experiments, and so forth.

 

      So, those are pretty much the recommendations in

 

                                                               173

 

      the guidance were very conservative.  So, that was

 

      the starting point.

 

                Now, quality-by-design thinking forces and

 

      challenges the industry to do this instead of FDA

 

      doing this, and it says simply that if you could

 

      understand your formulations and your manufacturing

 

      process to such an extent that you can start

 

      predicting the behavior of those things, then, you

 

      will start getting process understanding, and that

 

      information can allow you to document and justify

 

      what is critical, what is not critical for your

 

      given formulations instead of having a blanket peer

 

      guidance to say what is critical or what is not.

 

                So, that introduces the concept of needing

 

      to prove that hypothesis of your design space.

 

                With that in mind, what would that

 

      hypothesis be in the sense there are two aspects of

 

      the hypothesis that one could look at?

 

                One is proving that you have understood

 

      your formulation and manufacturing process to an

 

      extent that you can predict the behavior or its

 

      performance in terms of your shelf life, in terms

 

                                                               174

 

      of your bioavailability.

 

                So, these experiments that you conduct,

 

      you conduct them, you have to conduct them anyway.

 

      Instead of sort of approaching them as testing for

 

      the sake of testing, if you test those or you

 

      conduct those experiments as a hypothesis, then,

 

      you have a means to document your understanding and

 

      a means to, in a regulatory sense, prove your

 

      hypothesis through a hypothesis testing mode.

 

                So, when you think about it that way, the

 

      tests that you do today are no different except you

 

      are approaching those tests differently as

 

      hypothesis testing.

 

                What that does is that creates a

 

      flexibility for changing based on your

 

      understanding, based on what is critical to your

 

      formulation, and so forth.

 

                There were several challenges to that.

 

      One of the challenges was in terms of trying to

 

      prove your hypothesis, trying to do testing in more

 

      robust way, you do need to have estimates of

 

      variability and bring variability into discussion.

 

                So, yesterday, our discussion then focused

 

      on was a dissolution test procedure, which is a

 

      pivotal test procedure, which is a tool that is

 

                                                               175

 

      essential in product development.  The implications

 

      and the concerns FDA had in how are we setting

 

      specification, we saw a disconnect there.

 

                The variability in the dissolution test

 

      method may itself not be large, but the disconnect

 

      there was simply the suitability criteria opened a

 

      wider door than what our specifications are, and so

 

      forth.

 

                So, a stringent approach, a stringent

 

      mechanical calibration provides you a better handle

 

      on your target value or your mean values, and doing

 

      assessment of sensitivity of your formulations in

 

      that test system gives you an understanding and

 

      gives you a handle on the variability for your

 

      given formulation.

 

                So, that gives you a better handle on your

 

      variability, and that helps you start setting up

 

      your system to prove a hypothesis, and your

 

      hypothesis could be that my understanding of my

 

                                                               176

 

      formulations is such that I know what will happen

 

      to my shelf life if I change this or what will

 

      happen to the bioavailability if I change this.

 

                So, your stability program, your

 

      bioavailability studies that you do essentially are

 

      a confirmation of that. So, if we repeat that and

 

      use that as a decision criteria, you have become

 

      proactive.  So, that sets up the regulatory

 

      flexibility that is needed in the concept of design

 

      space.

 

                Similarly, I think bioequivalence is a

 

      hypothesis test.  Instead of approaching it just to

 

      document bioequivalence for the sake of documenting

 

      bioequivalence, you turn that around and say that

 

      is my test of hypothesis, I have understood my

 

      formulation, I have understood my manufacturing

 

      process, and I have also understood the product

 

      that I am duplicating or I am sort of reproducing

 

      to be equivalent, and therefore, my bioequivalence

 

      test now is the test of my hypothesis of how well I

 

      have understood, how well I have designed, and that

 

      opens a door for that test becoming a hypothesis

 

                                                               177

 

      test, confirming your knowledge base, and so forth.

 

                So, that combination opens the door for a

 

      scenario that Lawrence talked about, in a sense

 

      dealing with variability, which measurement system

 

      do we use to ask the right question.

 

                Clinical variability clearly is wider, and

 

      the drugs are approved on the basis of clinical

 

      trials.  The variability and quality has to be

 

      narrower by virtue of the system, and that is what

 

      Janet talked about in the sense variability and

 

      quality, or bioequivalence, we consider that as a

 

      quality test, not a clinical test, because it is

 

      not a clinical study, it's in healthy subject, is a

 

      confirmation that your variability is acceptable.

 

                With the question that we proposed with

 

      highly variable drugs is trying to understand what

 

      is the source of variability.  We know that many

 

      drugs are inherently variable because of the

 

      pharmacokinetic characteristics, metabolism, and so

 

      forth, that have nothing to do with the quality of

 

      the product.

 

                So, if you have understood your sources of

 

                                                               178

 

      variability in your manufacturing process, and your

 

      manufacturing process and your formulation

 

      strategies is consistent with the principles of

 

      formulation design that you have used previously

 

      and have documented lower variability, that gives

 

      us a handle to say that variability in this product

 

      is not expected to come from the product.  It is

 

      going to be inherent from the drug itself.

 

                Then, your biostudy becomes a test of

 

      hypothesis. Now, what is the hypothesis that we

 

      might want to test there?  The hypothesis could be

 

      test of means, an analysis of means rather than

 

      analysis of variance, because we have addressed the

 

      issue of variance in terms of being comfortable

 

      that the variance is coming from the drug

 

      substance, and we have enough confidence to say the

 

      variance for product is not expected to be

 

      different.

 

                So, instead of analysis of variance,

 

      analysis of means could be one option to consider

 

      there, instead of trying to do replicate design,

 

      tend to do large subject, and so forth.

 

                These are some of the initial thoughts

 

      that we have, and these are clearly not proposals

 

      at this time, and the intent of this discussion was

 

                                                               179

 

      to simply initiate discussion dialogue, to have the

 

      Pharmaceutical Committee get engaged in the

 

      discussion to see what opportunities we have.

 

                Highly variable drugs, topical products,

 

      we have been debating these issues for decades, and

 

      if we propose the same solutions, we will be

 

      debating those for the next couple of decades.

 

                So, I think it is an opportunity to think

 

      differently.  Similarly, I think some of the

 

      biopharm classification system in the context of

 

      design space, I think the biopharm classification

 

      system becomes a pivotal tool for your decision

 

      criteria that drives your decision to certain

 

      aspect based on the drug's property and what you

 

      are trying to achieve, and the biostudies that you

 

      do again become a confirmation of your hypothesis.

 

                So, one extension of BCS clearly is in the

 

      post-approval world, more so than the approval of a

 

      brand-new product, is the extension of SUPAC in

 

                                                               180

 

      application of design space.  So, that is the point

 

      that we tried to make.

 

                The aspect I think which is very difficult

 

      is industry often relies on us to tell them what to

 

      do, and it is very easy for them to have a check

 

      box.  FDA said this, let's do it, end of story,

 

      because the goal is to get the products approved.

 

                Well, that's one way of doing business,

 

      but in terms of I think FDA's role is to clearly

 

      ask the right questions.  As I again said at the

 

      training session, I think the decision system that

 

      we have for pharmaceutical quality is owned by the

 

      societies, not owned by the regulators or by the

 

      industry.  The decision is that of the society, and

 

      all of us are simply caretakers of that decision

 

      system.

 

                Unless we ask the right question, because

 

      of the scenario of the market failure, where the

 

      patient or the clinician cannot tell the difference

 

      between good quality and bad quality, the system

 

      may have inefficiencies built in, and even may not

 

      be asking the right question.  So, I think that is

 

                                                               181

 

      one of the things that we are trying to address.

 

                For the last four years, when we started

 

      the discussion on PAT, and so forth, clearly, the

 

      focus was on manufacturing, and relatively, I mean

 

      that was not an easy task, but relatively, that was

 

      easier to grasp for many because you already have a

 

      revolution of manufacturing that had occurred

 

      outside the pharmaceutical sector for the last 30

 

      years, and we are probably 30 years behind that

 

      revolution.

 

                Now, the most difficult part of the

 

      discussion, the journey starts now, is tackling the

 

      issue of science of design, and the reason I use

 

      the term "science of design" is that is a National

 

      Science Foundation terminology, which they have

 

      started a major funding program for focusing on

 

      science of design, because you cannot test quality

 

      in, and the infrastructure for U.S. in terms of

 

      design is so weak, especially in the software area,

 

      that is where the funding starts.

 

                Science of design provides you the

 

      scientific framework to say the empiricism that we

 

                                                               182

 

      have really, when structured, can provide you the

 

      fundamentals that you can start moving towards

 

      hypothesis-based decisions rather than just testing

 

      after the fact.

 

                So, I think that is the journey, and that

 

      is the first step in trying to think about it

 

      publicly.  For the last three, four years, or three

 

      years at least, our focus has been discussing only

 

      the first three bullets to a large degree.  It is

 

      focusing on supplements, focusing on deviations,

 

      focusing on testing and real-time release, and so

 

      forth.

 

                With ICH Q8, we started discussing the

 

      last two bullets, multiple CMC review cycles.  Why

 

      do we get into multiple CMC review cycles?  Because

 

      our reviewers are searching, trying to put the

 

      story together to see what are the issues as they

 

      try to approve this.

 

                Often, who gets blamed for the delay is

 

      the reviewers.  They are just trying to find the

 

      answers that they see to be comfortable in

 

      approving drug product.  If you get into multiple

 

                                                               183

 

      cycles, the reason is they don't have all the

 

      answers together.

 

                In Office of New Drug Chemistry, we are

 

      trying to move towards a quality oral summary as a

 

      starting point for this analysis.  Similarly, in

 

      OGD, we are moving towards a question-based review

 

      that will help sort of formulate the key question.

 

                But you have to keep in mind one aspect.

 

      That is, FDA does not develop or manufacture drugs.

 

      We are here to assess whether the quality is

 

      sufficient based on the standards that we have.

 

                Expecting FDA to give you the answers of

 

      how to develop and how to innovate and how to sort

 

      of make the case for science of design is expecting

 

      too much.  That is the reason why I think we wanted

 

      to sort of get the entire pharmaceutical community

 

      engaged to find and seek answers together rather

 

      than saying this is what we expect.

 

                So, that is the fundamental premise on

 

      which our discussions have been focused on, and

 

      clearly, I think industry has always argued for the

 

      last four years that it will increase the review

 

                                                               184

 

      cycle.  It will increase the delay over approval.

 

      We don't know how we will use it.

 

                One aspect of that argument simply says

 

      either they don't know what they are doing, they

 

      are afraid to hide that, or the concern is real.

 

      So, clearly, I think we understand that the

 

      concerns could be real, and that is the reason why

 

      I think we position the peer review process, but

 

      moving towards a quality system for review

 

      assessment, and so forth.

 

                But at the same time, I really think

 

      having the diverse disciplinary background that we

 

      have in the reviewer is an asset, not a liability.

 

      The challenge then is to construct the submission

 

      as a scientific submission hypothesis based.

 

                The key to that is without increasing the

 

      burden, utilizing the existing evaluations that we

 

      do, bioequivalence, stability, and others as part

 

      of hypothesis testing.  Suppose the hypothesis

 

      testing is your science that says I understood

 

      this, I expect this to happen, and here is the

 

      proof a priori providing that information.

 

                So, I think you have to think about that

 

      as a basis for discussion, particularly this

 

      morning's discussion, where we presented to you

 

                                                               185

 

      three examples, and the three examples essentially

 

      try to address the paradox that we run into, is

 

      trying to ascertain what is an acceptable

 

      variability, and trying to resolve the issue of

 

      whether our test procedures, whether our questions

 

      that we are asking are really reducing that

 

      uncertainty to get to an acceptable variability.

 

                Variability in the clinical, we are not

 

      touching, but that is what really we want to be

 

      lower or smaller than the clinical variability.

 

      So, our test procedures, and so forth, quote,

 

      unquote, I think gives us comfort that they are

 

      more discriminating, but the discriminating aspect

 

      of those test procedures is clearly an experience

 

      rather than science driven.

 

                So, how do you overcome the challenge, I

 

      think is the key issue.  So, in terms of

 

      formulation development, I really was fortunate in

 

      terms of getting trained in pharmacokinetics, as

 

                                                               186

 

      well as formulation and physical sciences, so that

 

      was my benefit.

 

                I can go across that and try to see both

 

      sides of it, and from my experience, I can see

 

      easy--not easy--approaches to sort of connecting

 

      those dots and aligning the current work that we do

 

      into a scientific structure.

 

                The challenge, the concern, a personal

 

      concern seems that is always with me is I think

 

      people who can connect all these dots are very few,

 

      and unless we build a team approach to this, we

 

      will be missing a lot of things, and much of the

 

      challenge today organizationally are the turf

 

      issues between different parts of the organization.

 

                We saw this very clearly with the biopharm

 

      classification system.  For Jack Cook to get the

 

      first submission in, he had to connect the PK

 

      Department, the Formulation Department, and so

 

      forth, and that was not easy, because each

 

      department is set in their own ways of doing

 

      business, their own test procedures, and the

 

      interfaces are difficult to manage, and much of the

 

                                                               187

 

      challenge today we have are dealing with the

 

      interfaces.

 

                So, the challenge is variability, what is

 

      an acceptable variability?  At the same time, the

 

      other challenge that we have is that of what is

 

      minimal expectation and what is optional

 

      expectation.  That is the definition, and that is

 

      the provision provided in ICH Q8.

 

                In some ways, this table, in my opinion,

 

      seems to give us a direction for trying to tease

 

      out what is the minimal expectation, what is an

 

      optional expectation.  Now, if you are a company

 

      with a generic or innovator, you will be making a

 

      tablet for the sake of argument.

 

                How much information does FDA really need

 

      to assess that quality was by design, and so forth?

 

      Actually, depending on how simple the dosage form

 

      is, things could be different.

 

                So, in the case of a conventional dosage

 

      form, what is the primary focus understanding the

 

      materials especially the new material that you are

 

      putting in existing materials, that is the drug

 

                                                               188

 

      substance into existing excipients, how well can

 

      you characterize that and how well can you predict

 

      the behavior in a set of mixtures of excipients

 

      which we have been using for about 150 years or so.

 

                We have been manufacturing hundreds of

 

      different formulations in the manufacturing

 

      process.  How can we capture that knowledge base

 

      and bring that?  So, if you bring that

 

      predictability and are able to do that, that amount

 

      of information that will be needed would be very

 

      minimal, and nothing probably more than what we

 

      have, but presented differently, that provides a

 

      way forward.

 

                But then you move towards more complex

 

      dosage form, I think where we don't have that, then

 

      even there, a science of design concept where you

 

      are testing hypothesis in a structured manner,

 

      leads you to a decision criteria.

 

                So, in this case, for example, I think our

 

      pharmaceutical quality characteristics of

 

      pharmaceutical equivalence, clearly, I think the

 

      minimal requirements are listed in this table, same

 

                                                               189

 

      active, identical amounts, same dosage form, route

 

      of administration.

 

                Same active, I think again how well can

 

      you characterize from simple to complex, there is

 

      an issue, identical amount, hopefully, that is not

 

      a challenge.

 

                Same dosage form raises many issues, and

 

      this is the nomenclature issue that we run into

 

      because we are dealing with nomenclature that

 

      started from Egypt--no, Egyptian based, no--our

 

      nomenclature, we have a lot of work to do in the

 

      nomenclature, because the description of our dosage

 

      form, and the performance and expectations, really

 

      have a lot of challenges built into that, and that

 

      is the source of constant legal issues that come

 

      about.

 

                That is the challenge to the pharmacy

 

      community, and just publishing the paper that Cindy

 

      published, the initial reactions from the

 

      reviewers, I was not surprised, but didn't see why

 

      it was important, so the pharmaceutical science

 

      community is ignorant of some of those issues.

 

                It is unfortunate because these things

 

      wrap us up in legal battles, and so forth.

 

                So, if you really look at this table, the

 

                                                               190

 

      acceptable variability that we have built into our

 

      system are our materials, our methods, and so

 

      forth.  That's the common cause variability that is

 

      part of the system, and simply the design aspect is

 

      how well you manage that and how well you make sure

 

      you don't introduce special causes.

 

                In terms of measurement system for

 

      bioequivalence, the key question is what is an

 

      appropriate measurement system and how you balance

 

      that with that of your sign that drives you to the

 

      right measurement system is the key and the

 

      goalpost for that.

 

                So, I think with that in mind, I think the

 

      premise that we had in putting this session

 

      together was the quality-by-design approach by

 

      pharmaceutical development can potentially provide

 

      an excellent means to address a number of

 

      challenges previously discussed and have been

 

      previously discussing for years.

 

                The topics of highly variable drugs, I did

 

      not see the discussion that occurred, that focused

 

      or was able to pinpoint a solution to that, and we

 

      didn't expect that.  That was not the purpose of

 

      it.  The purpose of the discussion was to get

 

      started.

 

                                                               191

 

                If you go back to the same old solutions,

 

      we will have the same old results.  So, how can we

 

      leverage this?  Is analysis of means, which the

 

      Japanese seems to have moved forward towards, a

 

      means to go forward?

 

                Again, it's a point of view right now that

 

      needs to be discussed, debated, and so forth, and

 

      how do we make a case.  Keep in mind, the way I

 

      look at bioequivalence, the way I look at stability

 

      studies that documents the shelf life, is a

 

      wonderful, final conclusion of a development

 

      report, which is a hypothesis test.

 

                In the regulatory setting, you need that

 

      level of clarity to make a decision, so what the

 

      pharmaceutical development does is provides you a

 

      means to come to that clarity, at that same time

 

                                                               192

 

      provide the right test procedures to create your

 

      hypothesis.

 

                So, that was the premise, and our hope was

 

      to engage you to start the journey together to see

 

      whether we are on the right track in our thought

 

      process, and the proposals or the discussion that

 

      we presented, clearly, is simply an initial thought

 

      of how should we proceed in even thinking about

 

      this.

 

                With that in mind, I will sort of pose the

 

      three questions to you.

 

                Help us structure our thought processes to

 

      discuss this in a structured way, to seek solutions

 

      that have eluded us for the last 10 or 15 years.

 

                DR. COONEY:  Thank you, Ajaz.

 

                Questions and comments from the Committee?

 

                Committee Discussion and Recommendations

 

                DR. SINGPURWALLA:  Yes, Ajaz.  There were

 

      several things you said, and I started making

 

      notes.  I think you were trying to say that

 

      hypothesis testing should be a basis for all forms

 

      of approval and all forms of activity, is that

 

                                                               193

 

      correct?

 

                DR. HUSSAIN:  I think it gives the

 

      structure--

 

                DR. SINGPURWALLA:  No, answer yes or no.

 

                DR. HUSSAIN:  Yes.

 

                DR. SINGPURWALLA:  No, I mean--

 

                DR. HUSSAIN:  Yes.

 

                DR. SINGPURWALLA:  Good.  A simple

 

      question to you.  How many statisticians do you

 

      have in your division, in your group?

 

                DR. HUSSAIN:  We don't have any.

 

                DR. SINGPURWALLA:  Well, the first

 

      suggestion is go hire one, because what you are

 

      really looking for is somebody who knows the art of

 

      testing hypothesis, and that is what statisticians

 

      do.

 

                So, I would strongly suggest that if

 

      that's the way you want to move, you should at

 

      least have some in-house experience.

 

                DR. HUSSAIN:  We have a whole department

 

      of biostatisticians.

 

                DR. SINGPURWALLA:  I know, but, you know,

 

                                                               194

 

      you ought to have your own lawyer and your own

 

      doctor, too.

 

                Now, you also said industry looks up to

 

      the FDA as to what to do.  Well, as a lay person,

 

      not connected with the pharmaceutical industry in

 

      any form, my sense is that the industry would

 

      rather wish you go away, but given that you are an

 

      approving organization, industry comes to you to

 

      make sure that their chances of getting approval

 

      succeed.

 

                DR. HUSSAIN:  True.

 

                DR. SINGPURWALLA:  That is just a general

 

      comment, but as far as your three bullets are

 

      concerned, I am very sympathetic to the

 

      presentation made by Lawrence demanding more

 

      information, and I heard the cross fire from my

 

      colleague, Paul Fackler, who was concerned, I

 

      quickly understand.  I fully support your thesis.

 

      How can more information hurt you?

 

                The question is how are you going to use

 

      that information judiciously.  Otherwise, you know,

 

      you will be loaded with eight volumes instead of

 

                                                               195

 

      the seven.

 

                As far as your second bullet is concerned,

 

      my suggestion is that is the whole premise of

 

      Bayesian inference, how to use data that is not

 

      directly observable on a certain phenomenon, but is

 

      auxiliary, to be able to make your tests of

 

      hypothesis.

 

                So, I can answer the second bullet by

 

      saying that pharmaceutical development information

 

      should be used and should be incorporated in

 

      whatever decisionmaking procedures you use through

 

      this particular inferential mechanism.

 

                That's all.

 

                DR. HUSSAIN:  Thank you.

 

                DR. COONEY:  Ken.

 

                DR. MORRIS:  One point that I wanted to

 

      bring up was that in talking to companies, to

 

      generic companies, there will be acknowledgment of

 

      the sort of cycle of questions.  When you talk to a

 

      lot of the innovator companies, they all say that

 

      they don't go through multiple CMC review cycles.

 

                DR. HUSSAIN:  Uh-huh.

 

                DR. MORRIS:  So, in that sense, the

 

      question that is often raised is so what do we get

 

      for this.  I know you have heard this, too.  I

 

                                                               196

 

      guess in partly response to Nozer's observation,

 

      which I think is spot on, is that they--"they"

 

      being the industry at large--wants FDA to

 

      disappear, in a sense, but not in reality.

 

                But what they do rely on, I guess, is FDA

 

      as a consultant as opposed to adopting the attitude

 

      that you would adopt if you were writing a

 

      scientific paper, which is here is my thesis, if

 

      you will, and here is my defense of it.  Now, you

 

      can judge whether my defense is sufficient.

 

                I think that is the mentality that has to

 

      shift, is that the industry has to say, now, look,

 

      I am not looking for FDA to tell me how to do this,

 

      I am going to do what I think is appropriate to

 

      make the case and defend the case scientifically,

 

      and then, having done this in a way that makes it

 

      hopefully more obvious to the reviewers, have them

 

      comment on the sufficiency of the application.

 

                I don't know if that is a comment or a

 

                                                               197

 

      question, but it's an observation in part.

 

                DR. COONEY:  Paul.

 

                DR. FACKLER:  I guess I should put on the

 

      record that we are not looking for FDA to

 

      disappear.

 

                [Laughter.]

 

                DR. FACKLER:  That's my hypothesis.  We

 

      will have to discuss working on that.

 

                Of course, I don't think we are looking to

 

      FDA to help us develop drugs.  7,000 drugs

 

      approved, somebody had on a slide over the past

 

      many years.  I think a large experience with those

 

      7,000 products to suggest that they are generally

 

      safe and efficacious.  I am certain there are

 

      exceptions to that, not a lot I don't believe.

 

                There is a handful of products for which

 

      we know how to develop what we consider to be

 

      bioequivalent and pharmaceutically equivalent

 

      products for which there doesn't seem to be a

 

      mechanism to get FDA to approve them, and those are

 

      the ones that I think we are really looking to FDA

 

      for guidance on, not how to develop them mind you,

 

                                                               198

 

      how to document appropriately that they are

 

      bioequivalent and pharmaceutically equivalent.

 

                We have obviously, records on all the

 

      pharmaceutical development activities.  They exist,

 

      so it is not as if we need to do more work to give

 

      those to you, and they are available for the field

 

      inspectors that come to our sites to do the

 

      preapproval inspections, and they often go through

 

      them.

 

                Really, the question in our mind is how

 

      will it help you here at the Center evaluate our

 

      applications, and how will you use those to help us

 

      demonstrate bioequivalence for this small fraction

 

      of products for which the standard analysis and

 

      treatment methods don't work.

 

                So, highly variable drugs is one of those

 

      classes. Topical products, we know how to get those

 

      approved, we know how to develop what are

 

      bioequivalent formulations.  I think the testing is

 

      a bit onerous, and revisiting it I think is a great

 

      idea.

 

                So, just some thoughts and I guess I will

 

                                                               199

 

      leave it at that.

 

                DR. COONEY:  Marv.

 

                DR. MEYER:  In sense, and in response to

 

      Nozer, I think that, in a sense, the FDA is almost

 

      like the group I have to deal with, the IRB.  I

 

      think the FDA is kind of they put the blessing on

 

      something, and now you have a shared marketing

 

      responsibility, a sharing of the guilt, if there is

 

      any to be shared.

 

                The IRB would ask questions.  They would

 

      reject protocols or they would accept protocols.

 

      You didn't like it when they were rejected, but

 

      sooner or later, you would get it approved.

 

                So, I think the industry probably enjoys

 

      having the partnership of FDA as long as the

 

      products ultimately get approved.

 

                I might rephrase your first bullet.  How

 

      can pharmaceutical development information help?  I

 

      would be more inclined to say what kind of

 

      information is needed to help extend the

 

      application, be it manufacturing, be it in vitro

 

      permeability or oral water partition coefficient,

 

                                                               200

 

      or whatever, some physiological, some--just don't

 

      restrict it to pharmaceutical development, because

 

      I think there are some other things we could put

 

      into play as a measurement and extend that waiver

 

      perhaps.

 

                Bullet No. 2, how can pharmaceutical

 

      development information be utilized?  I would say

 

      to make sure we have the best possible dosage form

 

      that one could make within reason before you even

 

      go to the clinic.  So, make sure that your--one

 

      example--make sure you don't have the six-vessel

 

      dissolution example.  Make sure that you understand

 

      the solubility and the PKA, and all of those

 

      physicochemical parameters.

 

                So, I think that kind of information, the

 

      idea being let's reduce the variability on the

 

      pre-body side, so that when we got on the human,

 

      all we have to do is worry about highly variable

 

      human beings.

 

                No. 3, I am not real sure how to tackle.

 

                DR. COONEY:  Pat.

 

                DR. DeLUCA:  I really commend your effort

 

                                                               201

 

      on the quality-by-design approach.  You know, this

 

      is a systematic approach to research and product

 

      development, and it must be hypothesis driven.  I

 

      think you are going to get that by establishing the

 

      decision tree, and this is going to be the plan.

 

                I think, you know, I try to encourage my

 

      students when they are going to do some research,

 

      is to map out what they are going to do and what

 

      they expect to get, and what they expect they get,

 

      they may get one thing they are expecting and one

 

      thing that they are not, and what are they going to

 

      do if they get that.

 

                So, to spend a little bit of time, even if

 

      it's a couple of days, planning, so that you have

 

      got a pretty good idea what it is you are trying to

 

      do.

 

                I think by your suggesting this to the

 

      industry, this quality by design, so you are

 

      expecting them to kind of carry it out.  So, I

 

      think this quality-by-design approach is a two-way

 

      street.

 

                I understand and appreciate what Paul is

 

                                                               202

 

      saying, but I think it's a two-way street, and you

 

      have got to be also available to be able to

 

      communicate to them what it is that you expect,

 

      knowing that they are carrying it out.

 

                But I think you can't just say, well, you

 

      know, you are doing the development, go ahead,

 

      don't ask us what to do, I think they have got to

 

      be able to ask, and you ought to be able to get

 

      some response to this.

 

                I think by carrying out the quality by

 

      design, and the decision tree, that, first of all,

 

      the variability in performance should not be due to

 

      the product.  You have pointed that out.  I think

 

      by going through this process, you will be able to

 

      assess the acceptable variability.

 

                You know, you have a highly variable

 

      product, maybe the limits can be widened.  Probably

 

      with a low variability product, you might be able

 

      to even tighten them.  So, those are the things.

 

                The BCS system is a very good tool.  I

 

      looked at that and I see it's a matter of

 

      solubility here.  That's all based on solubility. 

 

                                                               203

 

      You have II and IV that are low solubility drugs,

 

      and I think in your formulation efforts, the goal

 

      is to try to promote availability and

 

      bioequivalence.

 

                With the I and III, the high solubility,

 

      the goal is not to hinder availability or maybe to

 

      prevent bio-in-equivalence.

 

                So, I think the goal, the formulation

 

      strategy is going to be different where those drugs

 

      are in that classification, and I don't think you

 

      can get away, for all but the Class I, with an in

 

      vivo test.  I don't see how you can waive that for

 

      any of the other classes except I.

 

                DR. COONEY:  Art.

 

                DR. KIBBE:  This has been a fun couple of

 

      days, it always is.  Perspective.  To tell the

 

      industry that you do a really good scientific thing

 

      and send it to us, and that will speed up the

 

      process, I think might fall on deaf ears.

 

                The reason is that they have had

 

      experience sending stuff to the agency that wasn't

 

      exactly what the agency has been looking for, and

 

                                                               204

 

      it has taken iterations because part of the process

 

      of sending that kind of data is that you have to

 

      educate the people who are reading it to the value

 

      of it.

 

                So, when you get off of a guidance or a

 

      document that fits exactly what the agency has

 

      asked for, when you get off of that, no matter how

 

      good the science is, you have guaranteed yourself

 

      one more round, because there is going to be

 

      questions, and it is not that the agency is being

 

      mean or pejorative, it's that the people who are

 

      looking at it are going oh, wow, this looks really

 

      good, and maybe it's good and maybe it is really

 

      good, maybe it's not so good, boy, I would love to

 

      get them in here and talk about this, let's bring

 

      them in and let's talk about why they did that, and

 

      let's see what some of the background thinking is.

 

                There is no company that can put down all

 

      of that and write a textbook for it, and expect to

 

      do it on one time.  So, you are asking the

 

      companies to come in and help you develop what

 

      would eventually be guidances, and they are not

 

                                                               205

 

      really going to leap in there.

 

                I think this committee helps get us there

 

      in some ways, but you are going to end up having to

 

      write guidances for some of this stuff, and

 

      especially when you say give us pharmaceutical

 

      development information, because depending on the

 

      company, you are going to get different kinds of

 

      information.

 

                Different companies have divided their

 

      research and development efforts in different ways,

 

      and they will name it differently, and one company

 

      will give you a bunch of one kind of data and leave

 

      out a little bit of something else, so you really

 

      are going to, after we leave, leave you to your own

 

      devices, you are really going to have to come up

 

      with something that is a little bit more concrete.

 

                DR. HUSSAIN:  If I may, it is not exactly

 

      the way. We are planning our decision trees, and we

 

      will be developing the decision trees, and those

 

      will be the questions that we, as consumer

 

      advocates, will be asking.

 

                The rest of the job is through the

 

                                                               206

 

      industry, so we will provide a structure to this.

 

                DR. KIBBE:  The next part of where I was

 

      trying to go is that sometimes the questions that

 

      are asked are dependent on what kinds of decisions

 

      you are going to make.  A company might have a

 

      series of questions to ask if it wanted to design

 

      in quality.

 

                An academician might look at it and

 

      decide, ooh, wouldn't it be nice to know the

 

      mechanism rather than just know how to control it,

 

      and regulation is really aimed at knowing that

 

      whatever you are controlling is going to get me

 

      consistent quality, and those are different kinds

 

      of questions, and all worth asking and worth

 

      knowing about.

 

                I think we have another topic to come up

 

      with, which is the research end of it, and that

 

      kind of feeds into that.  It would be really nice

 

      to be able to have a collaborative research effort

 

      with academia and industry and the agency on these

 

      issues that is not going to necessarily be the

 

      answer, but a place to ask really good questions.

 

                Then, of course, the last thing I wanted

 

      to say is it is impossible to read people's minds,

 

      so you don't really know what they are thinking

 

                                                               207

 

      when they get to that point.  You know, it's hard.

 

                DR. COONEY:  Tom.

 

                DR. LAYLOFF:  As we were visiting earlier,

 

      I think that there is enough knowledge base in the

 

      industry and academia for formulation and

 

      manufacturing to produce products which dissolve

 

      and which are uniform without failure.

 

                I think what we see in all of these,

 

      especially No. 1 and 2, is wrestling with an

 

      unknown, and how do you evolve a waiver around

 

      things that are not well defined and unknown.

 

      Certainly, highly variable drugs belong with drugs,

 

      but they don't fit in the BCS, because they are not

 

      understood well enough.

 

                The BCS takes a rough cut at physical

 

      properties of the substance, but not at the

 

      transport mechanisms or the metabolic processes

 

      that might control some of the properties of the

 

      drug.

 

                So, I think that the waiver is going to be

 

      hard for broad-brush strokes, but maybe narrow

 

      categories can be trivialized to a few physical

 

      properties.  The highly variable drugs are

 

      startling, and I don't know where you go, but I

 

      think they are the class that sort of makes you

 

                                                               208

 

      uncertain about the waivers and the BCS.  They put

 

      an uncertainty in there also.

 

                The last one, therapeutic equivalence of

 

      topical products, I don't know what to do with

 

      that.

 

                DR. HUSSAIN:  There is an aspect, Tom, I

 

      think, if I may, the Class IV drug inherently tends

 

      to be more variable, so there is a relationship

 

      between variability and class, I think.  Hopefully,

 

      when Raman's database is audited and ready, I think

 

      you might see a pattern there, because I think

 

      variability, physiologic variability--I actually

 

      was going to show a slide that we just finished,

 

      our analysis of a Class IV drug that Raman sent me

 

      over the weekend.  You will shocked at the

 

      variability that we see.

 

                But there is a mechanism to sort of start

 

      identifying what is the source of variability with

 

      the GI physiology, with its metabolism, and so

 

      forth, so you can actually start thinking about a

 

      structure to say what characteristics make the drug

 

      more variable.

 

                So, in some ways, I think the BCS

 

      classification, and this is a proposal of Les

 

      Benet, is to extend that.  I mean he has simply

 

                                                               209

 

      used that to extend and start predicting the class

 

      of metabolism of those things.

 

                So, I think we could consider sort of

 

      characterizing the sources of variability and see

 

      if we can start.

 

                DR. LAYLOFF:  Have you ever tried to put

 

      another column on the box, the BCS box, like

 

      polarizability of the molecule or footprint of the

 

      molecule, geometry?

 

                DR. HUSSAIN:  No.  What Lawrence actually

 

      has done is actually went back to the structure and

 

      predicted the bioavailability, so there is an

 

      element of that.  We recently published a paper on

 

                                                               210

 

      going back and actually predicting the permeability

 

      from the structure, so there is an element of that.

 

                Professor Les Benet has now extended that

 

      to actually a classification system to include

 

      metabolism.  So, that is a recent proposal.  To

 

      there is lots of progress in that area.

 

                DR. LAYLOFF:  So, that will fit into the

 

      possibility of going towards a waiver business.

 

                DR. HUSSAIN:  I have not studied some of

 

      the latest ones, so I just have seen the papers,

 

      but not studied them, so I can't say, but I think

 

      there are some positive signs there.

 

                DR. COONEY:  Ken.

 

                DR. MORRIS:  Tom made actually one of my

 

      points better than I would have probably, but to

 

      that point, I think if you look at BCS-3, clearly,

 

      from the data we saw, I was a little surprised that

 

      that was the highest variability class, as we

 

      talked about earlier, but if you include some of

 

      the work that Les has been doing, was that

 

      editorial?

 

                If you include some of the work that Les

 

                                                               211

 

      has been doing, so that you could subdivide Class

 

      III in particular into mechanistic subcategories,

 

      if you will, it certainly seems like there ought to

 

      be something in Class III, that is some element or

 

      some subcategory of Class III that should be ripe

 

      for waiver.

 

                I mean if their premise is valid, the

 

      hypothesis is valid, then, there ought to be a way

 

      to do that.

 

                The other thing with respect to what a lot

 

      of people have talked about, Jerry before, and Paul

 

      and Art to some extent, when I was in industry,

 

      which was admittedly a while ago, we used to

 

      generate what in my particular case we called IDSC,

 

      initial drug substance characterization report.

 

                As we were talking about earlier, these

 

      exist already in most places.  The only question I

 

      think, or only caveat I guess that needs to be

 

      added to that would be that once you are to the

 

      point of filing, in the light of what you know post

 

      this initial drug substance characterization

 

      report, you might truncate what you provide only

 

                                                               212

 

      based on what is or isn't necessary.  It's not like

 

      you want to just provide everything.

 

                But in many instances, and I have seen

 

      this in consulting, these documents almost exist in

 

      place already, and it is just really a question of

 

      pulling the right things out or adding what is

 

      relevant in.  I think that is relatively common.

 

                DR. HUSSAIN:  Again, I do know they exist,

 

      and that was the reason for starting this, because

 

      I knew that was already there, but it was bringing

 

      those into making decisions, because I think Moheb

 

      mentioned we get volumes and volumes and volumes of

 

      things that we have to sort through, which is not

 

      value added.

 

                We get supplements after supplements,

 

      which is not value added.  So, the whole idea is to

 

      utilize that and make the decision, and then

 

      without having to get all of those things that we

 

      have to sort through, and so forth.  So, that is

 

      one way of looking at it.

 

                DR. COONEY:  Michael.

 

                DR. KORCZYNSKI:  Just a few generalized

 

                                                               213

 

      comments relative to communicating to the industry.

 

      As this unfolds and is described to industry, I

 

      think they ought to be reminded that I see the

 

      quality-by-design plan for pharmaceuticals somewhat

 

      analogous to the Center for Radiological Health and

 

      Devices' developmental design plan, and it is sort

 

      of a similar concept, and they ought to be made

 

      aware that indeed there is an analogous situation

 

      here.

 

                The other thing is I think back when we

 

      talk about industry not knowing what the FDA wants,

 

      well, there was a climate back in the 1980s

 

      relative to sterilization technology of sterile

 

      products, and a number of companies were concerned

 

      when they were making submissions and were

 

      receiving some rejections and questions, gee, what

 

      does the FDA want.

 

                Well, the FDA went on to draft a guideline

 

      for sterilization technology information when

 

      submitted in NDAs, and actually went on several

 

      performances at different cities, about four in

 

      all, and that was discussed openly with industry,

 

                                                               214

 

      and that became very effective and really did a lot

 

      in terms of dispelling some misunderstandings.  So,

 

      a similar approach could be undertaken here at some

 

      time.

 

                DR. COONEY:  Any additional comments from

 

      the Committee?

 

                The request here for this topic comes,

 

      well, as on the screen, these are our initial

 

      thoughts, are we on the right track.  As you can

 

      see, there are three questions that have been

 

      posed, I think somewhat rhetorical questions in

 

      that you are not looking for a vote on these

 

      particular issues.

 

                I have tried to capture what I think is

 

      the consensus of what people have been saying, and

 

      if you will allow me to try and summarize this

 

      point, and the question I want to ask the Committee

 

      is does this--what I am going to say--does this

 

      capture what we collectively have said.

 

                I am looking for omissions in this summary

 

      and I am also looking for things that shouldn't be

 

      there, so that is the input I am looking from the

 

                                                               215

 

      Committee, and then I will ask if we, in general,

 

      agree that this is a consensus.

 

                The platform here is that there is a need

 

      for a better understanding of the science of

 

      formulation design will lead to improved product

 

      quality with reduced variability.  In fact, this is

 

      the foundation for quality by design.

 

                That the implementation of quality by

 

      design will require additional information on the

 

      product development process.  This is what has been

 

      generally referred to as the product development

 

      report, and that the FDA wants to use this

 

      information, this product development report, which

 

      is ill-defined at the moment, or loosely defined at

 

      the moment, to do three things, as you have

 

      outlined in your earlier slides:

 

                To extend the BCS-based waiver for

 

      immediate release products, to facilitate approval

 

      of highly variable drugs, and to facilitate the

 

      establishment of pharmaceutical equivalence of

 

      topical products.

 

                So, those are the three general goals, and

 

                                                               216

 

      from this conversation, there is a need that has

 

      been discussed to work with both the industry and

 

      the reviewers, in other words, the system needs to

 

      be receptive to receive and use effectively new

 

      information, and that it is important to add

 

      clarity on what information is required, as well as

 

      how that information will be used, for instance, to

 

      establish a bioequivalence, and that the general

 

      consensus seems to be to recommend that the FDA

 

      continue down this path to address quality by

 

      design and define its use to facilitate issues in

 

      the regulatory approval of drug products.

 

                DR. HUSSAIN:  A point of clarification.

 

                DR. COONEY:  Please.

 

                DR. HUSSAIN:  One aspect also I think,

 

      please consider this, the CTD Q B2 section has the

 

      sections and everything defined.  The ICH Q8

 

      defines what information goes where, and so forth.

 

                So, in that sense, it is already

 

      structured, it is already part of the guideline,

 

      and one of the aspects, the timing of this meeting

 

      in relation to ICH also has to be considered here,

 

                                                               217

 

      because starting this Saturday/Sunday, we are

 

      moving towards putting together decision trees for

 

      the dosage forms in Q6A, so you will see the train

 

      leaving the station of how these decision trees

 

      will evolve, and the goal is to get to Step 2 by

 

      2006.  So, that process is beginning next week.

 

                Now, this meeting, one of the other

 

      aspects is also is we focused this on the generic

 

      side for one reason, also was we often have

 

      hesitation as generics really do not have the level

 

      of involvement there, because there are just

 

      observers there.

 

                So, I think is also a plea for the

 

      generics to keep engaged with that process, because

 

      that process is leaving, and Europe already has

 

      their decision trees for all of this, and we are

 

      not fully happy with that.  We want to make sure

 

      the decision trees that evolve the next six, seven

 

      months will be the science base.

 

                DR. COONEY:  I think that in the

 

      presentation that you have made, Ajaz, the role of

 

      decision trees at multiple points in this process

 

                                                               218

 

      was clarified, so I believe that that is clear.

 

                I would ask the Committee, did this

 

      summary capture what you believe was the essence of

 

      the conversation from this morning and this

 

      afternoon?

 

                DR. SINGPURWALLA:  I think in terms of

 

      proper vocabulary, I think the word "hypothesis"

 

      was constantly used by Ajaz.

 

                DR. COONEY:  I noticed that it was used.

 

                DR. SINGPURWALLA:  I also noticed that it

 

      was eliminated from your summary, and so I am just

 

      trying to remind you whether you want to endorse it

 

      or not.

 

                DR. COONEY:  I did not leave it out by

 

      design.

 

                DR. SINGPURWALLA:  It was because of

 

      variability.

 

                DR. COONEY:  It was a bit of variability.

 

                DR. SINGPURWALLA:  I would try to give a

 

      strengthening hand to Ajaz and Helen and all in

 

      terms of endorsing what they want to do by

 

      specifically including that in the vocabulary, so

 

                                                               219

 

      that it succinctly conveys the intention of what

 

      they want to do.

 

                I wouldn't say anything about the fact

 

      they don't have any statisticians in their group.

 

                DR. COONEY:  This will get written up in

 

      minutes of the meeting, and I will see to it that

 

      it is appropriately worded and we will check the

 

      vocabulary and the grammar.

 

                DR. SINGPURWALLA:  Spelling.

 

                DR. COONEY:  Does everyone--so, the train

 

      is leaving, you should be aboard the train and

 

      rolling down the tracks, and I believe it is fair

 

      to establish the expectation that probably at our

 

      next meeting, we will hear something more

 

      definitive.

 

                DR. HUSSAIN:  At least on some aspects of

 

      that.

 

                DR. COONEY:  Yes.  It would be nice to

 

      have all the problems solved by the next meeting.

 

                I think this brings us to closure of this

 

      topic. We are going to go to another topic which

 

      deals with the research, and I would suggest that

 

                                                               220

 

      we take a five-minute stretch break and reconvene

 

      at approximately 7 minutes past 2:00.

 

                [Break.]

 

                DR. COONEY:  I realize that a five-minute

 

      stretch break is a very short period of time, but I

 

      wanted to look around and see those who didn't get

 

      up and stretch, and that I would go wake them up.

 

                DR. KIBBE:  Just so that we keep

 

      everything on point, stretch breaks are an

 

      extremely highly variable process.  If you say 5

 

      percent in stretch breaks, that could mean anywhere

 

      from 10 to 40 minutes.

 

                DR. COONEY:  I said 5 minutes, and there

 

      has already been a 2-minute variation on that, so

 

      your point is well made, Art.  I will take that

 

      into account in the future.

 

                The next topic is a very important one and

 

      it revolves around the Criteria for Establishing a

 

      Working Group for Review and Assessment of OPS

 

      Research.

 

             Criteria for Establishing a Working Group for

 

             Review and Assessment of OPS Research Programs

 

                DR. HUSSAIN:  The topic is a request to

 

      form a subcommittee and this advisory committee to

 

      have a peer review process for research.  We

 

                                                               221

 

      already have a committee under CBER and we need to

 

      migrate that committee under this to have a process

 

      in place for Office of Biotechnology Research

 

      Program.

 

                But also I think we want to take this

 

      opportunity to put in a place for a peer review for

 

      all of our research programs in one umbrella.  With

 

      that in mind, all we seek today from you is an

 

      endorsement to form a subcommittee and define the

 

      scope and charter as the committee gets formed, and

 

      so forth.

 

                But for you, we have just a presentation

 

      of the background of what the current system is,

 

      and then Keith will come back and ask the

 

      questions.

 

                DR. COONEY:  The first presentation will

 

      be by Kathleen.

 

           CBER Peer Review Process for Researchers/Reviewers

 

                DR. CLOUSE:  I have been asked to put

 

                                                               222

 

      together a simple but concise summary of how we do

 

      peer review for the OBP Research Program.

 

      Hopefully, you can follow along without much

 

      confusion.

 

                I have divided this up into four

 

      discussion topics.  The first is an outline of the

 

      Researcher/Reviewer Model.  Secondly, how

 

      Researcher/Reviewer Program is monitored.  The

 

      process for external scientific review.  Then, the

 

      promotion and conversion evaluation or PCE

 

      Committee through which the researcher/reviewers

 

      are converted to permanent positions and promoted

 

      through the GS system.

 

                The Researcher/Reviewer Model is something

 

      that has existed at CBER for a while.  We do use it

 

      in OBP.  We have individuals who do both research

 

      and review, and we also have individuals that do

 

      full-time review.  So, before I describe the

 

      program, I would like to emphasize the fact that

 

      more than 75 to 80 percent of our full-time

 

      reviewers have come up through the

 

      Researcher/Reviewer Program.  So, this is also in

 

                                                               223

 

      part a training program for our reviewers.

 

                The responsibility of a

 

      researcher/reviewer, first of all, is to conduct

 

      research that is relevant to the FDA mission, and

 

      this research is generally dealing with specific

 

      products, and that can be for mechanism of action,

 

      for toxicity, or surrogate measures of efficacy.

 

                It can be related to product classes,

 

      specific diseases, or therapeutic modality, and it

 

      can also be associated with the development of

 

      methods and standards by which products can be

 

      prepared.

 

                In addition to the research, the

 

      researcher/reviewer performs regulatory review, and

 

      this is at the level of investigational new drug

 

      applications, as well as biologic license

 

      applications, and they also are involved in

 

      conducting inspections for specific BLAs.

 

                They also contribute to policy development

 

      as they become more senior in the structure.

 

                The funding of OBP research, the majority

 

      of the funding is provided at the OBP level from

 

                                                               224

 

      our operating funds, and it is distributed on a per

 

      capita basis.  A portion of the allocation is held

 

      aside, and these additional funds are distributed

 

      by OBP based on research prioritization.

 

                The research prioritization is determined,

 

      not just at the office level, but from guidance at

 

      the agency level and what the agency deems to be a

 

      priority for that given year.

 

                We also have access to competitive funding

 

      through the CDER Review Science and Research or RSR

 

      program.

 

                We have obtained competitive funds through

 

      the Office of Women's Health granting program.

 

                We also have access to competitive funding

 

      through the NIH Intramural Grant Program.  These

 

      are limited for the most part for research dealing

 

      with AIDS and, more recently, for

 

      counterbioterrorism efforts.

 

                We also have some funds that is obtained

 

      through CRADAs and inter-agency agreements.

 

                The program monitoring is done at multiple

 

      levels. The first tier of monitoring is done by the

 

                                                               225

 

      Lab Chief.  Now, the Lab Chief generally has their

 

      own research program and is responsible for several

 

      additional principal investigators.

 

                The Lab Chief does not determine the

 

      research focus, however, they do assess the

 

      research productivity of the principal

 

      investigators and offer some guidance if they don't

 

      appear to be productive enough, if they are spread

 

      too thin, and so on.

 

                But more importantly, the Lab Chief is

 

      involved in the actual training of the principal

 

      investigators and any Staff Fellows working under

 

      them on the regulatory review process, and the Lab

 

      Chief evaluates the ability of the individuals to

 

      perform regulatory review.

 

                The next tier for evaluation is the

 

      Division Director, and the Division Director

 

      discusses the scientific productivity and

 

      regulatory abilities at least twice a year, but

 

      this is often done through the Lab Chief, because

 

      they have the first tier of evaluation.

 

                The third level is at the Office Director

 

                                                               226

 

      and Associate Director for Research level.  Here,

 

      the scientific productivity is assessed via

 

      publications.  What is taken into consideration is

 

      not just the number of publications, but the type

 

      of journals, the impact factor, and also the

 

      relevance to the FDA mission.

 

                Finally, we also have External Scientific

 

      Review or site visits.  The purpose of the External

 

      Scientific Review is, first of all, to determine

 

      the relevance of the research program to the FDA

 

      mission; secondly, to evaluate research

 

      productivity; third, to assess the regulatory

 

      contribution, and this is a portion of the External

 

      Review, and also to provide input regarding

 

      resource allocations.

 

                In general, the input is we should get

 

      more resources, but there is usually not much the

 

      agency can do about it.

 

                The External Scientific Review ideally

 

      occurs every four years.  Now, the research group

 

      that is reviewed, generally, it is all principal

 

      investigators within a specific research lab, in

 

                                                               227

 

      other words, you have the Lab Chief and whichever

 

      principal investigator works in that particular

 

      lab.

 

                On some occasions, we have grouped site

 

      visits based on expertise, so that the site visit

 

      reviewers don't have to be duplicated or they can

 

      overlap in their review process.

 

                The Site Visit Committee, and this is most

 

      important and one reason that we need to bring up

 

      the issue with this advisory committee today, the

 

      Chair of the Site Visit Committee is generally a

 

      member of the parent advisory committee.

 

                Previously, this was the Biological

 

      Response Modifier Advisory Committee, however,

 

      since our transfer from CBER to CDER, they have

 

      renamed the group and refocused the emphasis of the

 

      committee members.  That is currently known as the

 

      Cell, Tissue, and Gene Therapy Advisory Committee.

 

                So, what is happening is as individuals

 

      with expertise in our area end their term, they are

 

      being replaced by individuals with more of a focus

 

      on cell and gene therapies.

 

                In addition to the Chair, there are one or

 

      two external scientists with relevant research

 

      experience or expertise for each principal

 

                                                               228

 

      investigator under review.  When possible, if there

 

      is a member of the Advisory Committee that has the

 

      relevant expertise, they also will be asked to

 

      serve on the Site Visit Committee.

 

                The format of the external scientific

 

      review, first of all, the committee is assembled,

 

      they are given the review package, and they review

 

      the scientific program in a formal setting.

 

                At the conclusion of the site visit, a

 

      summary is given to the Center and Office

 

      Directors, so that they have a pretty good idea of

 

      how the site visit went.

 

                Several weeks later, a preliminary written

 

      report is sent to the Center for review, and then

 

      that preliminary report is presented by the Chair

 

      of the Site Visit Committee to the Advisory

 

      Committee, and the report is ratified by the

 

      Advisory Committee before it can be used by any of

 

      the scientists.

 

                A copy of the official report is then

 

      provided to the Center and Office Directors, and

 

      individual reports are given to scientists under

 

      review.

 

                The site visit report is used in the

 

      following manner.  Within two years of the site

 

                                                               229

 

      visit, a favorable report can be used for tenure or

 

      conversion of the principal investigator to a

 

      permanent position.

 

                Within four years of the site visit, if

 

      the individual is already tenured, it can be used

 

      for promotion to a GS-14 or 15, and these

 

      promotions are permanent.  It can also be used as

 

      supporting documentation for internal grant

 

      applications or external grant applications when

 

      applicable.

 

                Now the Promotion and Conversion

 

      Evaluation or PCE Committee actually makes the

 

      decisions on the conversion and promotion of

 

      scientists.  So, the purpose of the committee is

 

      the conversion of Staff Fellows to tenured Civil

 

      Service research and regulatory positions.  They

 

                                                               230

 

      also are involved in promoting tenured Civil

 

      Service research and regulatory scientists to the

 

      next grade level, as I mentioned before.

 

                The composition of the committee is as

 

      follows.  There are two tenured principal

 

      investigators from each of the Research and Review

 

      Offices, so we have our own two representatives

 

      from the Office of Biotechnology Products. That is

 

      myself and also Emily Shakter from the Division of

 

      Therapeutic Proteins.

 

                In addition, there are members from the

 

      Office of Blood Research and Review, the Office of

 

      Cell, Tissue, and Gene Therapy, and the Office of

 

      Vaccines at CBER.  Each of those respective offices

 

      has one full-time ad hoc reviewer, so for any one

 

      situation, one of those ad hoc full-time reviewers

 

      also serves on the committee.

 

                There is a representative present from the

 

      Office of Personnel Management to make sure all the

 

      procedures are followed, as needed, and as legal,

 

      and there is one representative from the CBER

 

      Office of the Center Director.

 

                The guidances that are used by the PCE

 

      Committee for Promotion and Conversion, there is a

 

      general CBER guide for the evaluation of research

 

                                                               231

 

      and regulatory scientists from GS-13 to GS-15,

 

      which is available on their website.

 

                There is also a Research Grade Evaluation

 

      Guide, and this is from the General Schedule

 

      Position Classification Guide, which I think is

 

      from the Office of Personnel Management.

 

                Generally, through the use of these

 

      guidances, there is a scoring system, and the

 

      scoring for promotion and conversion is actually

 

      documented by the Office of Personnel Management,

 

      and that is maintained and brought back for

 

      comparison for each subsequent promotion

 

      opportunity.

 

                There is additional information that is

 

      requested by the PCE Committee.  This includes a

 

      publication summary, as well as a presentation

 

      summary, and these can be scientific presentations,

 

      as well as regulatory presentations.

 

                You have to include a summary of your

 

                                                               232

 

      regulatory work, as well as examples of your

 

      regulatory reviews.  You have to have a copy of

 

      your external scientific review report, and you

 

      also need letters of recommendation from experts

 

      outside FDA that are familiar with the

 

      investigator's research.

 

                Now, the use of the site visit and the PCE

 

      Committee systems has advantages and disadvantages,

 

      and this is the slide I will end with.

 

                The current advantages are that scientific

 

      and technical positions are evaluated by scientists

 

      who are actually familiar with the activities

 

      performed, and the scientific community is expected

 

      to have greater confidence in decisions made by

 

      peer scientists.

 

                The current disadvantages to the existing

 

      system, first of all, is the cost to OBP, OPS, and

 

      CDER.  Each site visit costs us not just for

 

      bringing the scientists in, but also because the

 

      administrative office overseeing the site visit is

 

      the CBER Division of Scientific Advisors and

 

      Consultants, so we also pay for their time and the

 

                                                               233

 

      coordination of the site visits.

 

                It is also difficult to coordinate site

 

      visits across the two Centers at this point.  As I

 

      mentioned before, there is a change in expertise of

 

      the Advisory Committee.  Members of the Advisory

 

      Committee have been instrumental in advising us

 

      with regard to the scientific expertise that is

 

      needed to review the biologic therapeutic

 

      applications.  We really would be remiss if we

 

      didn't have that input from the external

 

      scientists.

 

                There is also a difference in the

 

      regulatory workload among the members of the PCE

 

      Committee, and this has to do with different

 

      structures of the offices.  One of the offices has

 

      a structure similar to ours, where the product

 

      reviewer does both research and regulatory.

 

                Two of the other offices actually have a

 

      structure where there is a separate division that

 

      does the majority of the review, and the scientists

 

      are viewed more as consultants, so the workload

 

      varies.

 

                There is also a difference in regulation

 

      of BLAs versus INDs.  Very often in the Office of

 

      Vaccines and the Office of Cell, Tissue, and Gene

 

                                                               234

 

      Therapy, the majority of the applications are under

 

      IND, and they don't have the experience of dealing

 

      with as many biologic license applications as we

 

      do, so there is some disconnect with evaluating the

 

      actual amount of the work.

 

                There are also differences--and this is my

 

      latter point--in the systems for performing review,

 

      and that has to do with whether the

 

      researcher/reviewer has both the full product

 

      review or CMC review responsibilities and

 

      inspections.

 

                DR. COONEY:  Thank you.

 

                Some questions?  Tom.

 

                DR. LAYLOFF:  How many people are involved

 

      in the review side of this group?

 

                DR. CLOUSE:  I think it is split about

 

      50-50.  Keith can answer that.

 

                DR. WEBBER:  Within the office, there is

 

      about 14 full-time reviewers, and there is about 36

 

                                                               235

 

      or so who are research/reviewers, who are supposed

 

      to spend half their time doing review and half-time

 

      research, but I think they spend more time review.

 

                DR. LAYLOFF:  The laboratory people, where

 

      are they located?

 

                DR. WEBBER:  They are all physically

 

      located down in Building 29A and B of NIH campus.

 

                DR. LAYLOFF:  And the full-time reviewers

 

      are located?

 

                DR. WEBBER:  They are located in the same

 

      place. They work together and share meetings.

 

                DR. LAYLOFF:  So, there is 50 people

 

      there.  There is 36 and 14.

 

                DR. WEBBER:  Right, plus there are support

 

      staff and technicians who work in the laboratories

 

      for the research program.

 

                DR. LAYLOFF:  How many applications or

 

      supplements do they review per year?

 

                DR. WEBBER:  Applications, we get

 

      generally around four full field applications per

 

      year, and that varies.  Sometimes we have gotten up

 

      to nine.  We get about between 150 and 200

 

                                                               236

 

      supplements per year, somewhere in that range, and

 

      then annual reports.  INDs, we have approximately a

 

      little over 400 products in IND.

 

                DR. LAYLOFF:  Thank you.

 

                DR. SELASSIE:  I have a couple of

 

      questions.  You said the majority of the funding is

 

      provided at the OBP level.  Could you tell me how

 

      these funds are appropriated to each department, is

 

      it a peer review process, do they write proposals,

 

      how those decisions are made?

 

                DR. CLOUSE:  No, it is divided, as I

 

      mentioned, on a per capita basis to each of the

 

      programs, a portion of the money, and then there is

 

      a portion that is held back for research

 

      prioritization, which is awarded based on

 

      productivity or the nature of the research and how

 

      it fits in with the current prioritization for FDA.

 

                DR. SELASSIE:  I suppose scientific merit

 

      comes in there someplace.

 

                DR. CLOUSE:  The scientific merit pretty

 

      much comes in the site visit process and the annual

 

      review of productivity.  So, yes it does.  We

 

                                                               237

 

      haven't had an instance that I can recall where we

 

      have had any lack of productivity.  I mean

 

      generally, the thought is the resources would

 

      diminish for someone who is not productive, but we

 

      haven't come across that in the years that I have

 

      been there.

 

                DR. SELASSIE:  I assume that most of the

 

      Lab Chiefs basically supervise the labs, and they

 

      don't do any research, they supervise the PIs under

 

      them?

 

                DR. CLOUSE:  No, we do research.

 

                DR. SELASSIE:  They do research, too?

 

                DR. CLOUSE:  Yes.

 

                DR. SELASSIE:  So, how much of your time

 

      is like spent during research and how much on

 

      review activities?

 

                DR. CLOUSE:  I was asked that question at

 

      a presentation at NIH last week, and my Staff

 

      Fellow said 200 percent was regulatory.  I would

 

      say more than 90 percent of my time right now is

 

      spent on regulatory, and what I do researchwise is

 

      done at home.

 

                DR. SELASSIE:  When you do this, the

 

      reviews of your researchers/reviewers, and I guess

 

      go to the various steps, to the advisory committee.

 

                                                               238

 

      At some point, I guess eventually, the reviewer

 

      gets to see the individual report. At some time can

 

      they respond to issues that were raised in those

 

      reports before they go on file as, you know, done?

 

                DR. CLOUSE:  Currently, that is a touchy

 

      issue.  In general, what has happened in the past,

 

      we get a draft report and if the report is not

 

      consistent with what happened at the summary

 

      meeting, at the level of the Center Director, you

 

      know, the individuals under review or their

 

      immediate supervisors are not allowed to contact

 

      the Advisory Committee members.

 

                It is not considered appropriate.  But if

 

      there is an issue, you know, potentially, at the

 

      level of the Center or Office Director, they can

 

      contact the Committee Chairperson, and they would

 

      deal with it at that level.

 

                DR. WEBBER:  Generally, an effort has been

 

      made to try to maintain, since we have a

 

                                                               239

 

      multi-tiered review process, to maintain a site

 

      visit as independent as possible, so that there

 

      isn't any--unless, as Kathleen said, if there is a

 

      serious issue, serious problem with the review that

 

      comes from the Site Visit Committee, that they may

 

      have been biased or something like that, we

 

      generally try to avoid getting involved before the

 

      report is made final.

 

                But afterwards, certainly, if there are

 

      issues or concerns or additional information, that

 

      the Site Visit Committee didn't have in hand at the

 

      time, that can be added to the review process.

 

                DR. SELASSIE:  One other question.  In

 

      choosing outside reviewers, does the reviewer, the

 

      person under consideration, do they have a choice

 

      or do they give you a list of outside reviewers,

 

      and can you pick from them?

 

                DR. CLOUSE:  Generally, the person from

 

      the Scientific Advisers and Consultants Division

 

      asks for a list of names.  This list of names

 

      cannot be anyone that you have collaborated with or

 

      a friend.

 

                That list of names is provided to the

 

      Chair, whoever has been identified as the Chair of

 

      the Site Visit Committee.  They are not obligated

 

                                                               240

 

      to choose anyone from those, however, once they do

 

      choose your reviewer, they do let you know or

 

      contact the individual principal investigator and

 

      ask if there is any conflict or problem with who

 

      has been chosen.

 

                So, you have some say in the process, but

 

      you don't have the final decision.

 

                DR. SELASSIE:  One last thing.  You talked

 

      about the cost to OPB of bringing in outside

 

      reviewers.  Have you ever thought of doing

 

      videoconferencing?  I know the EPA does that.

 

                DR. CLOUSE:  For site visits?

 

                DR. SELASSIE:  Yes.

 

                DR. CLOUSE:  We haven't pursued it at this

 

      point, but then again, we are in the process--

 

                DR. WEBBER:  It's something we can

 

      consider, but oftentimes it's an all day affair,

 

      because you have a meeting in the morning with

 

      presentations from each of the people under review,

 

                                                               241

 

      and then there is discussions within the committee,

 

      and usually, it takes pretty much all day.

 

                We might save some money by bringing

 

      people in by video as opposed to in person, but we

 

      would probably lose a great deal in terms of the

 

      actual interaction.

 

                DR. CLOUSE:  It is pretty much like you

 

      have with the interaction of the Advisory

 

      Committee.  You would lose a lot if everybody

 

      teleconferenced in consistently.  It is just a

 

      little more fluid if you have the people there.

 

                The one thing I did forget to emphasize,

 

      and that is, for the researcher/reviewer, when you

 

      do get an application in, whether it's an IND

 

      original submission or biologic license

 

      application, or supplement, that's a priority, your

 

      research stops.

 

                So, very often when you look at someone's

 

      productivity--and this is one reason why it's

 

      difficult to assess productivity, let's say,

 

      annually, your productivity can go like this

 

      depending on what your regulatory workload has been

 

                                                               242

 

      for any given year.

 

                DR. SELASSIE:  Thank you.

 

                DR. COONEY:  Carol.

 

                DR. GLOFF:  Just a couple of quick

 

      questions.  So, there are 14 people who are

 

      full-time reviewers, 36 who are half and half,

 

      which we know it's not really 50-50.  Are there

 

      people who just do research?

 

                DR. CLOUSE:  Only technical staff, and if

 

      we have funding for postdoctoral fellows, those

 

      individuals do full-time research.

 

                In the majority of cases, if the

 

      postdoctoral fellow is a citizen or has a green

 

      card, and expresses an interest in doing the

 

      regulatory, the next step for them is to become a

 

      staff fellow.  Then, very often, lately, the

 

      majority of our staff fellows have gone on to

 

      become full-time reviewers.

 

                DR. GLOFF:  Then, my other question is--I

 

      know we are going to have a presentation by Dr.

 

      Collins--I am just curious.  Setting aside the

 

      Center for Biologics, and obviously your group now,

 

                                                               243

 

      but are there other review centers or sections in

 

      the other review centers that have a process

 

      similar to what you just described?

 

                DR. CLOUSE:  If I recall correctly, NCTR

 

      is the other center that has a structure similar to

 

      ours.

 

                DR. GLOFF:  I guess I don't think of them

 

      as being a review center, but I may have that

 

      wrong.

 

                DR. WEBBER:  I am not sure if CVM has a

 

      research review program, I don't know about that,

 

      but that will be something to look into.

 

                DR. HUSSAIN:  I think CDRX, CFSAN, they

 

      have research programs.  They are not, as Keith

 

      said, reviewers, and they do have aspects of this,

 

      but not in the form that exists under CBER right

 

      now.

 

                DR. COONEY:  Let's proceed on with Jerry

 

      Collins and the next part of the presentation.

 

      Then, we will have a chance to come back for more

 

      questions.

 

                       CDER Peer Review Research

 

                DR. COLLINS:  Good afternoon.  The

 

      background document that I prepared focuses more on

 

      the review of research programs than on review of

 

                                                               244

 

      individual scientists, but when I realized that we

 

      were going to be covering both topics, you will see

 

      the copies that you have of my slides, I tried to

 

      cover both.

 

                In my time at FDA, in addition to working

 

      in CDER, I have been asked from time to time to

 

      help other centers evaluate their research

 

      scientists or their programs, so, in general, I

 

      would say that the systems for peer review of

 

      individuals in all centers have more similarities

 

      than they have differences.

 

                One of the mentors, Bob Dedick, used to

 

      say that biologists are always looking for

 

      differences, and engineers are always looking for

 

      similarities, so this may just reflect my

 

      engineering background.

 

                Every employee at FDA has a semiannual

 

      management review, so that is the baseline review.

 

      Everything else is built upon that.  Within the

 

                                                               245

 

      Center for Drug Evaluation and Research,

 

      non-laboratory scientists, the people do full-time

 

      review and policy work, have promotion letters to

 

      the 14 and 15 through things that we call expert

 

      reviewers or master reviewers.

 

                Those committees are composed of internal

 

      FDA members and they are intended for non-managers

 

      to have a promotion letter.  Those promotions,

 

      unlike in our laboratory side, are permanent, and

 

      they are not periodically recertified.

 

                We have a few additional personnel system,

 

      Title 42 and Title 38, that are used for non-lab

 

      personnel and at least until recently they have not

 

      been subject to committee review or

 

      recertification.

 

                Finally, Congress created the Senior

 

      Biomedical Research Service that I will be talking

 

      about that at FDA, is implemented to cover both

 

      non-laboratory and laboratory scientists.

 

                Within CDER, to go back to being a

 

      biologist and to highlight the differences between

 

      the way the OBP Committee has been set up through

 

                                                               246

 

      the CBER system versus the CDER system, we have

 

      always had a requirement of a minimum of 50 percent

 

      to the voting members of our committees to be

 

      scientists from outside our Center, not outside our

 

      research program, not outside our office, but they

 

      have to be outside CDER.

 

                Most often, for convenience and for

 

      compliance with some of the nuances of personnel

 

      review regulations, we have used individual

 

      scientists from NIH and frequently from other

 

      centers, and our staff has also served on the

 

      review committees of other centers.

 

                Their purview is the hiring or promotion

 

      of scientists to GS-14 or GS-15, and effectively,

 

      they are three-year renewable promotions, they are

 

      not permanent.  A survey of the record indicates

 

      that very few people are not renewed, but

 

      occasionally, it has been a leverage to use when a

 

      person unexpectedly underperforms.  We point out to

 

      them that at their next review, this will be noted.

 

                The Senior Biomedical Research Service is

 

      an agency-wide program.  We have an agency-wide

 

                                                               247

 

      Credentials Committee supplemented by external

 

      consultants, and it covers promotions from GS-15 to

 

      essentially above the regular Civil Service pay

 

      scale, and it clearly has a recertification

 

      requirement, so it is either four or five years

 

      depending on the center that the people come from.

 

                That is a recertification with teeth, and

 

      I can't discuss individual cases, but plenty of

 

      discussions are made, and it has provisions, for

 

      example, for a one-year renewal instead of a

 

      four-year renewal to keep your feet to the fire if

 

      necessary.

 

                Some laboratory scientists are also

 

      covered by Title 42, and as I said, for the non-lab

 

      folks, up until recently there have been no

 

      committees or recertification associated with that.

 

                Again, I think based on my experience in

 

      consulting for other centers, as well as my

 

      experience within CDER, I think there you could

 

      nitpick some of the differences across the review

 

      of individual scientists.  I think generally, there

 

      is a lot more similarities, and it is a lot more

 

                                                               248

 

      effective, understood by all parties, and truly an

 

      ongoing process.

 

                But as I said in my backgrounder, the peer

 

      review of research programs themselves has

 

      considerable polarity, and so as we just heard in

 

      the discussion after the previous speaker, CBER and

 

      NCTR have lab research programs that are much more

 

      like the academic or the NIH model, site visits

 

      conducted by advisory committees.

 

                We have five other laboratory-based

 

      research units at the FDA:  Center for Devices and

 

      Radiological Health, Center for Food Safety and

 

      Applied Nutrition, Center for Veterinary Medicine,

 

      the field organization in CDER, and for lack of a

 

      well-defined term, I call that a corporate or a

 

      management model, very similar to what a

 

      pharmaceutical company does for its research

 

      programs.

 

                The primary evaluation of research

 

      programs is internal by the program management.

 

                Within CDER, we have had occasional

 

      episodic external review.  They are not formally

 

                                                               249

 

      established, they are not regularly conducted.

 

      Sometimes they are conducted when a problem is

 

      noted by center management in their semiannual

 

      review.  Sometimes it's just we haven't done one

 

      for a while, let's do it again.

 

                I think what we are looking for today in

 

      terms of advice from the committee is whether that

 

      is really the most effective model.

 

                In terms of the ad hoc reviews that we

 

      have had, the FDA Science Board, which is another

 

      advisory committee like this one, usually picks one

 

      topic at a time, and might review the program, for

 

      example, in genetics or genomics across all the

 

      centers, rather than just a genomics or genetics

 

      program at CDER.

 

                The predecessor of this committee is

 

      called the Generic Drug Advisory Committee, and it

 

      started the tradition of at least having some site

 

      visit-like character and certainly information

 

      briefings.

 

                So, last October I was here in front of

 

      this committee talking about the OTR research

 

                                                               250

 

      programs.  I did that also back in March of 2003.

 

      We have training sessions periodically.  I think

 

      those are very valuable in helping to orient you

 

      folks and to prepare you to give advice to us, and

 

      we do get feedback, but that is not the same in any

 

      stretch of the imagination as a formally organized

 

      peer review process.

 

                The ad hoc reviews from external folks of

 

      CDER programs usually are problem solving

 

      exercises.  At one time, we had another advisory

 

      committee called the Antiviral Drug Products

 

      Advisory Committee, still have it, and when we

 

      created a laboratory program on antiviral drug

 

      products, we made it part of the charter of that

 

      committee to conduct periodic reviews of that

 

      laboratory.

 

                Unfortunately, that laboratory did not

 

      flourish, and that laboratory no longer exists and

 

      has been abolished. So, our review process does

 

      have teeth, it is not just a friendly pass among

 

      colleagues.

 

                We also had, in the Center for Drug

 

                                                               251

 

      Evaluation, support of a cardiovascular

 

      pharmacology laboratory, and after an ad hoc

 

      external review, that laboratory was decided to be

 

      no longer funded.

 

                So, again, reviews aren't as frequent and

 

      regular, and don't capture the benefit of that, but

 

      they do provide sort of a final chance to prove

 

      yourself when things are going bad.

 

                Internal reviews.  I mentioned a little

 

      bit about this at a training session earlier this

 

      week.  There is a tradition of annual presentations

 

      to the Center Director, the Deputy Center Director.

 

      A year might take longer than 12 months to call it

 

      an annual review, but that has been a goal for a

 

      long time.

 

                I think we have had a number of serious

 

      efforts to have a Research Coordinating Committee,

 

      and it is always important when evaluating Center

 

      for Drug Evaluation and Research, is that we are

 

      not primarily about laboratory programs.  There is

 

      at least as much, or perhaps more, research that is

 

      conducted outside the laboratory.

 

                Things like reviewing files to find common

 

      class effects of drugs, things like creating

 

      databases to improve the review process, thinking

 

                                                               252

 

      about looking at new standards for either safety or

 

      efficacy.

 

                So, all these programs in a very large and

 

      rambling center like ours is hard to keep track of,

 

      so the Research Coordinating Committee currently,

 

      in its current form, is chaired by the Deputy

 

      Center Director, is an attempt to try to pull

 

      together centerwide databases of research and to

 

      help center management in a pretty tough decision

 

      of resource allocation.

 

                Now, I made a comment at the training

 

      session that the good news is we have high level

 

      visibility with the Deputy Center Director as our

 

      chair.  The bad news is because at that level, the

 

      person is so busy, we get a lot of cancellations.

 

                Shortly after that, all our Blackberries

 

      went off and another meeting was scheduled.  I

 

      don't think there is any connection, but I am

 

      nervous.

 

                Within OPS, for example, one example of

 

      the kinds of non-laboratory research that is

 

      conducted by the Informatics and Computational

 

      Safety Analysis Staff, looking at

 

      structure-activity relationships, and spinning off

 

      various databases in terms of different elements of

 

                                                               253

 

      safety, carcinogenesis, reproductive genetox, and

 

      the like.

 

                Elsewhere, the Biostatistical Biometrics

 

      Office, the Office of Drug Safety, Office of

 

      Information Management, and Office of Clinical

 

      Pharmacology and Biopharmaceutics have

 

      well-established research programs.

 

                Funding has come up in the question and

 

      answer period.  The primary sources of funding for

 

      CDER research programs, as with the tradition and

 

      history of OBP when they were in CBER, and now that

 

      they are part of CDER, is primarily determine by

 

      office management.

 

                Managers should be accountable for the way

 

      they spend all their dollars, whether it is for

 

      review, policy development, travel, or research,

 

                                                               254

 

      and that has been the policy in CDER as long as I

 

      have been here.

 

                In addition, there is, as was mentioned in

 

      the OBP presentation, there is an opportunity to

 

      get funds from outside your individual budget

 

      through the Review Science and Research program,

 

      but those funding areas are limited to certain

 

      areas determined by CDER management.

 

                There is an internal peer review by CDER

 

      scientists.  Almost always those funds are not

 

      intended for laboratory-based research, but they

 

      are intended to foster primarily activities, such

 

      as database generation within review divisions.  In

 

      a sense, they are equivalent of a laboratory.

 

                Within the agency, we have a number

 

      offices, the Office of Health Science Coordination

 

      chaired by the Deputy Commissioner for Science, and

 

      the Office of Women's Health, have had a certain

 

      amount of money set aside every year over the last

 

      10 years, and competition agencywide for these

 

      funds is conducted.  The priorities, again, the

 

      categories that they are willing to fund are set in

 

                                                               255

 

      advance, so that is the management of the funding

 

      office.

 

                Proposals are peer reviewed both for

 

      quality and for relevance to the priorities that

 

      have been set by the funding office.  The peer

 

      review is conducted only by internal FDA staff, but

 

      both lab and non-lab proposals are accepted.

 

                Within our Laboratory Research Program, we

 

      do have CRADAs, cooperative research and

 

      development agreements, which is a source of

 

      outside money.  We also have had occasional

 

      interagency agreements with other federal agencies,

 

      primarily NIH.

 

                As you can imagine, those sources of funds

 

      come with strings attached to them.  By law, they

 

      can only be spent on the purpose defined in the

 

      CRADA or in the interagency agreement, so there is

 

      a compromise between what might be your primary

 

      mission and the mission of your partner in those

 

      agreements.

 

                I would say, in summary, that I grew up

 

      scientifically, mostly within the NIH system, so I

 

                                                               256

 

      am used to a system of peer review, site visits.  I

 

      see a lot of strength in that.

 

                I am also a very strong proponent as a

 

      part-time manager, part-time scientist, of holding

 

      management accountable.  I don't think it is

 

      necessarily bad to make it a part of the review of

 

      management on how well or how poorly they fund

 

      research and how well or how poorly they evaluate

 

      it.

 

                I am very strongly convinced of the nature

 

      of applied research, relevant research.  I probably

 

      have a reputation for pushing that angle too hard.

 

      Whenever I do, I try to remember my own interview

 

      when I was joining FDA, and I had a chance to

 

      interview with Commissioner Frank Young.

 

                He took me in his office and he said,

 

      well, Jerry, I see you come from an applied

 

      background and I am glad to see that because that

 

      is what you should be doing in CDER. So, I thought,

 

      great, I can really depend on the Commissioner to

 

      support this, whenever anybody is off doing blue

 

      sky stuff, I will just tell them what the

 

                                                               257

 

      Commissioner told me.

 

                But he didn't finish his sentence there.

 

      He looked out his window and he said, "You see that

 

      sky.  I think your job description ought to include

 

      an element that says you spend 20 percent of your

 

      time looking out the window or at least doing blue

 

      sky research."

 

                So, I think applied research, directed

 

      regulatory relevance, is clearly doable, a little

 

      bit of flexibility in terms of pursuing something

 

      that may not quite be there from a regulatory

 

      relevance, but has at least some hope in the

 

      future, not 80 percent, but I will take

 

      Commissioner's Young's 20 percent.

 

                I was asked by some of the other members

 

      of the Office of Testing and Research to mention

 

      that we do occasionally very short-term projects, a

 

      little bit more of a testing flavor than of

 

      research.  They are given by Office of New Drug

 

      Chemistry of Office of Generic Drugs, or another

 

      Office of Pediatrics, one of the other offices

 

      within CDER.

 

                They have a very short turnaround time,

 

      and in a sense they are peer reviewed, because the

 

      offices are either pleased with our work product or

 

                                                               258

 

      they are not, and if they are not, they don't come

 

      to see us again.

 

                That really is my view of how the CDER

 

      research operation has worked up until the time

 

      that the merger occurred and OBP joined us.

 

                Committee Discussion and Recommendations

 

                DR. COONEY:  Thank you, Jerry.

 

                Are there some questions from the

 

      Committee?  Cynthia.

 

                DR. SELASSIE:  Just one question.  You

 

      mentioned that the funds for research are usually

 

      used for database generation at CDER?

 

                DR. COLLINS:  That is in the particular

 

      category of review science and research, so the

 

      primary applicants, in fact, the principal

 

      investigator for all those things has to be a

 

      primary reviewer.  Other people can be

 

      co-investigators.  So, by its nature, the funds

 

      that are available there tend to be used for

 

                                                               259

 

      creating databases.

 

                At the training session earlier this week,

 

      I talked about the project that we were

 

      co-investigators on, looking at a review of

 

      neuropharmacology NDAs over the last 10 years.

 

      That was an example of a project that was funded by

 

      the Review Science and Research program.

 

                What does it do?  In theory, it gives a

 

      little bit of release time to the reviewers.  In

 

      practice, they probably just do it on top of their

 

      regular job.  It gives us some travel money.  The

 

      poster that came out of that meeting, it was

 

      presented at a meeting in Florida.  The travel

 

      funds came out of the RSR budget.  Publication

 

      expenses come out of that budget.

 

                DR. SELASSIE:  But could the Informatics

 

      people help you all with that?

 

                DR. COLLINS:  The Informatics people are

 

      sort of built into the process.  If you are going

 

      to make a database generally available at FDA, you

 

      have to start by talking to the people who run

 

      either the OIM or the OIT, the Information

 

                                                               260

 

      Management Information Technology.  All databases

 

      have to fit the new corporate model, so that is

 

      there.

 

                If in that process, they find out there is

 

      some off-the-shelf software that is available for

 

      it, that's great.  Our Neuropharm project has used

 

      Microsoft Access, it wasn't one of these

 

      mega-databases.  The hard part was extracting the

 

      data from paper documents that had come in over the

 

      last 10 to 25 years and putting them in electronic

 

      format.

 

                MS. WINKLE:  Let me make it clear, too,

 

      these aren't large sums of money.  They may be

 

      $150,000 that are put aside, or may be up to 250

 

      depending on what the budget allows each year, and

 

      the amounts allotted are usually like in small

 

      amounts, 5- or $10,000 just to keep a project

 

      going.  It is not enough to do any real bench

 

      research on.

 

                DR. LAYLOFF:  How many people are we

 

      talking about, Jerry?

 

                DR. COLLINS:  The Office of Testing and

 

                                                               261

 

      Research has about 70 laboratory-based persons.

 

      That includes a handful of support staff.  We don't

 

      actually have a classification system that permits

 

      us to tease out the technical support staff from

 

      the principal investigators.

 

                So, when we were talking about the numbers

 

      for OBP being 36 review-based scientists, we

 

      actually have, in all, 65 review-based scientists

 

      or something like that, but many of them would be

 

      classified as technical support under a different

 

      classification system.  So, we just lump all our

 

      folks together.

 

                DR. LAYLOFF:  You don't have a review

 

      function corresponding?

 

                DR. COLLINS:  There are no line reviews

 

      assigned. There are probably 10 of our staff who

 

      spend more than 25 percent of their time doing

 

      review and policy work.  I chair several committees

 

      that are related to the writing of guidance

 

      documents, the FDA-wide Imaging Initiative, which

 

      is primarily a non-lab operation.

 

                Up until this year when these other duties

 

                                                               262

 

      took me away, I did a tertiary review of every

 

      single new molecular entity that was submitted to

 

      the Center for Drug Evaluation and Research, an

 

      average of about 30 a year.

 

                Other scientists, like John Strong, work

 

      in the Drug Metabolism, Drug Interaction area,

 

      picked up the slack as I have moved into other

 

      areas.  On the Chemistry side, we have folks who

 

      are consultants to that process, but we don't have

 

      signature authority on the review of any product

 

      from within--

 

                DR. LAYLOFF:  There is no growth concept

 

      like people going from research and moving into

 

      doing some review, and then becoming reviewers full

 

      time?

 

                DR. COLLINS:  Well, Tom, we call that a

 

      "stealing away" phenomenon, not growth.  Let's be

 

      clear that Ajaz Hussain was the Director of our

 

      Division of Product Quality, and Moheb Nasr was the

 

      Director of our Division of Pharmaceutical

 

      Analysis, and on and on, like that, so it is a

 

      measure of quality and desirability, but you have

 

                                                               263

 

      to debate the idea that it's growth.

 

                [Laughter.]

 

                DR. COLLINS:  Just kidding.

 

                DR. COONEY:  I think it is probably

 

      appropriate as the next step, Keith, if you would

 

      pose the question that you would like to Committee

 

      to address.

 

                DR. WEBBER:  I think what we are looking

 

      for here is not really a lot of discussion about

 

      setting a peer review program or a site visit

 

      program or the pluses or minuses of the various

 

      aspects of it, but really just to come to the

 

      Committee to look for an agreement to support the

 

      creation of a subcommittee that will help us to

 

      develop the criteria and the processes within OPS

 

      to evaluate the research programs, the diverse

 

      research programs that we have within the office

 

      now.

 

                Certainly, you can ask questions in that

 

      regard, I think hopefully, we can come to an

 

      agreement that you would be interested in that,

 

      because we are interested in that.

 

                DR. COONEY:  I would like to open up this

 

      question for discussion.  Our charge this afternoon

 

      is not to solve this problem, but rather to ask if

 

                                                               264

 

      we concur in making a recommendation towards the

 

      establishment of this committee.

 

                Tom?

 

                DR. LAYLOFF:  So, you would basically

 

      create a subcommittee, which would come back and

 

      report to ACPS on the activities?

 

                DR. WEBBER:  That would probably be one

 

      avenue or one aspect of developing a subcommittee,

 

      and most subcommittees do come back to report to

 

      the Committee, so I would imagine that would be how

 

      it would work.

 

                DR. LAYLOFF:  So, the ACPS then would be

 

      providing guidance to the subcommittee on how to

 

      proceed with this?

 

                MS. WINKLE:  Let me point out, too, if you

 

      determine to create a subcommittee, you would have

 

      to have two members of the Advisory Committee that

 

      would serve on this subcommittee, so not only would

 

      you expect for the subcommittee to come back and

 

                                                               265

 

      report to the Committee, the Committee would also

 

      have input into the subcommittee through those two

 

      members.

 

                DR. COONEY:  Marv.

 

                DR. MEYER:  Well, I don't have any

 

      fundamental objection to that approach.  I just

 

      wonder why it is necessary.  You have several

 

      models out there in different groups in FDA

 

      already.

 

                It would seem like you could use some of

 

      their expertise, as well as some of your own

 

      people, to develop your own criteria for promotion

 

      and for funding and research, and not have folks

 

      like us sitting around the table and muck it all up

 

      for you.

 

                DR. WEBBER:  I don't know that we are

 

      necessarily looking for that much in terms of

 

      mucking with the process.

 

                [Laughter.]

 

                DR. WEBBER:  But I think that right now we

 

      have a system within OBP's site review system,

 

      which I think clearly needs to be replaced, and we

 

                                                               266

 

      have a system which is now with OTR in the same

 

      office, and I think if we can look at both aspects

 

      of both research programs, and try to come up with

 

      something that works for both, it doesn't

 

      necessarily have to be the same, but has to be

 

      something that will be consistent within the Office

 

      of Pharmaceutical Science, and provide us with some

 

      external guidance on the research programs that we

 

      have, something that we have always gotten from--or

 

      at least with OBP, we have gotten that from the

 

      CBER system, but we need to move forward.

 

                DR. COONEY:  I would like to thank Marvin

 

      for introducing technical terminology into the

 

      minutes.

 

                Art.

 

                DR. KIBBE:  I think it's a wonderful idea

 

      that you have a review of research activities

 

      within the agency, and if you need our help, then,

 

      we should stand ready to do that.  So, you lay it

 

      out and we will populate the committee for you.

 

                DR. DeLUCA:  I agree.  I think you have

 

      certainly given this some thought, and you think it

 

                                                               267

 

      is important.  I certainly think you do need some

 

      external input into this, into the research that is

 

      being performed and the caliber of it.

 

                You do, I guess, derive some funding from

 

      NIH, too, right, for this?

 

                DR. WEBBER:  Funding from NIH to establish

 

      a peer review?  No.

 

                DR. DeLUCA:  No, no, for the research.

 

                DR. WEBBER:  For the research, no, we get

 

      money from NIH for the research.  We get money from

 

      operating funds, we get money, as you saw, from

 

      other sources.

 

                DR. DeLUCA:  Maybe we ought to tap that

 

      source.

 

                DR. CLOUSE:  There is money from NIH, but

 

      only through Intramural NIH grants, so the money we

 

      can apply for is limited.  I mean we have a number

 

      of investigators who have been funded through the

 

      Intramural AIDS programs, and received AIDS grants.

 

                We have received money for equipment

 

      through that program, and this year NIAID has also

 

      started an Intramural Grant program for

 

                                                               268

 

      counterbioterrorism research, and we have people

 

      who have applied for that.

 

                So, we have been successful.  That is

 

      competitive with the other NIH institutes, and we

 

      have received funding.

 

                DR. COONEY:  I think the fact that there

 

      are different models to look at, both models that

 

      exist within the system now as a consequence of the

 

      merger, as well as alternative models and practices

 

      from other organizations, it would very interesting

 

      and useful to look at.

 

                Perhaps--not perhaps--I am sure there are

 

      best practices from alternative models, and I would

 

      hope that this working group would be able to reach

 

      outside and look at a number of alternatives and

 

      come up with recommendations towards a system that

 

      is most appropriate for the diverse activities that

 

      are present here.

 

                DR. WEBBER:  I agree with that completely.

 

      I don't think we need to reinvent the wheel

 

      entirely, but to look at what other systems are in

 

      place, and take the best practices from those.

 

                DR. COONEY:  I personally see this as an

 

      important activity for a working group.

 

                If there are no further questions from the

 

                                                               269

 

      Committee, what I would like to do is to pose this

 

      as a recommendation.  I think it is appropriate for

 

      us to vote on it, which I will go around the table

 

      and ask for votes in just a moment.

 

                The alternative votings are yes, no, or

 

      abstention.  What we are voting, this would be a

 

      recommendation by this committee to the FDA to form

 

      a working group of the ACPS to address the criteria

 

      and processes for evaluating the OPS research

 

      programs.

 

                We are now empowering the creating of a

 

      committee, but we are recommending that they go

 

      forward with the formation of a subcommittee.

 

                Marv?

 

                DR. MEYER:  A point of clarification.  You

 

      mean a subcommittee under ACPS with two members

 

      from this group, not members of this group.

 

                DR. COONEY:  Yes, that is what I meant.

 

      Thank you.

 

                Since we began with Art yesterday on the

 

      previous vote, we will begin with Tom today, and I

 

      would like to go around and have a yes, no, or

 

      abstention.

 

                Tom.

 

                DR. LAYLOFF:  Yes.

 

                                                               270

 

                DR. COONEY:  Cynthia?

 

                DR. SELASSIE:  Yes.

 

                DR. SWADENER:  Yes.

 

                DR. COONEY:  Mike.

 

                DR. KORCZYNSKI:  Yes.

 

                DR. COONEY:  I think I heard yes from

 

      both.

 

                Morris?

 

                DR. MORRIS:  Yes.

 

                DR. COONEY:  Pat?

 

                DR. DeLUCA:  Yes.

 

                DR. COONEY:  Carol?

 

                DR. GLOFF:  Yes.

 

                DR. SINGPURWALLA:  Yes.

 

                DR. KIBBE:  Yes.

 

                DR. MEYER:  Yes.

 

                DR. COONEY:  And yes for myself.  We have

 

      a unanimous 11 yes's, zero no's, zero abstentions,

 

      and 11 yes's total to 11 votes.

 

                Thank you very much, Keith.  This brings

 

      this topic to a close.

 

                I would just like make two further

 

      comments before I believe we close for this

 

      session.  One, I appreciate very much the very

 

      thoughtful contributions that each of the

 

                                                               271

 

      presenters have made in a very nice style.  I like

 

      the data-driven presentations, and the fact that

 

      they were very concise.

 

                I would like to thank the Committee

 

      members for their very, very thoughtful comments on

 

      all of the topics that allowed us to move I think

 

      reasonably efficiently through what was a very full

 

      and very important agenda.

 

                This is an important committee to OPS as

 

      it helps them as they go forward and craft an

 

      aggressive agenda being proactive and changing some

 

      of the paradigms with which they work.

 

                I would certainly like to ask the

 

                                                               272

 

      question, if anyone on the committee has thoughts

 

      on things that we should be talking about, should

 

      be addressing that we haven't talked about before,

 

      this is an open-ended question. We can deal with

 

      thoughts that you might have now, but it is meant

 

      to be a permanent question on the table, that as we

 

      have things that we think should be addressed by

 

      this committee, I would hope that people would be

 

      very forthright in bringing them up, so that we can

 

      come back to them in an ongoing manner.

 

                Does anyone have any thoughts at the

 

      present time?

 

      Nozer.

 

                DR. SINGPURWALLA:  Not on the question you

 

      asked, namely, but I would like to make some

 

      comments about the format of the meeting.  I would

 

      like to suggest that the number of presentations be

 

      cut down and the length of each presentation be cut

 

      down, and there be more time for discussion

 

      instead.

 

                This puts not only less burden on the

 

      committee, but it also puts less burden on the

 

                                                               273

 

      staff and yourselves.  I know you work very, very

 

      hard, and I know this is a stressful thing for you

 

      to do, and it is not as stressful for us, but I

 

      think cutting these down would be of some value to

 

      us.

 

                DR. COONEY:  Any other comments or

 

      thoughts from the Committee?

 

                If there are no objections, I will call

 

      the meeting to a close and thank you all very

 

      much--oh, I am sorry.  Ajaz, Helen, please.

 

                     Conclusion and Summary Remarks

 

                DR. HUSSAIN:  Let me quickly summarize.

 

      Just wanted to sort of encapsulate some highlights

 

      that were, in my opinion, I think the key

 

      directives and recommendations we heard from the

 

      Advisory Committee.

 

                First, I think I would like to acknowledge

 

      and thank our colleagues from Health Canada who

 

      have attended this session and have shared with us

 

      their perspective as the meeting went along, and

 

      also shared their experience on the same issues.  I

 

      think we share quite a bit in common and we are

 

                                                               274

 

      collaborating on many fronts.

 

                The meeting started with a discussion on I

 

      believe an important topic where the tactical plan

 

      that we proposed in some ways is a paradigm shift,

 

      and that is the reason we proposed a tactical plan

 

      instead of proposing putting forward a proposal to

 

      you, because this will allow the community to go

 

      back and debate and vigorously engage in this

 

      topic, so that when we come back with a proposal,

 

      we hope the entire community will connect to that.

 

                The accomplishments there were, in a

 

      sense, nothing new from a quality sense from

 

      outside the pharma sector, but I think we

 

      introduced some of the tools and methodologies that

 

      have been utilized and approaches that have been

 

      utilized successfully in other industries to

 

      attempt to move towards a more probabilistic

 

      approach to setting specifications that allow us to

 

      be risk based and science based, and bring a high

 

      level of ability to manage variability in

 

      measurement systems in the case of dissolution, but

 

      more so I think start to focus on the product of

 

                                                               275

 

      interest and see how we can take that information

 

      and move towards a controlled philosophy that

 

      allows us to gauge the capability of a process and

 

      also the state of control.

 

                I believe that will be extremely important

 

      for our CMC reviewers and GMP inspection staff to

 

      really connect, because establishment of state of

 

      control and ability to have confidence in that is

 

      the part of continuous improvement, and that is one

 

      way of reducing the need for supplements, and so

 

      forth.

 

                So, that becomes a basis for moving

 

      forward, but at the same time, dissolution is just

 

      one of the physical performance attributes of

 

      interest.  This becomes a model for all other

 

      specifications especially with respect to physical

 

      attributes.

 

                The challenges are even greater on the

 

      other fronts including particle size, and so forth.

 

      So, I think the strong endorsement of the Committee

 

      really sends a strong signal and provides the

 

      support that we needed to really push ahead with

 

                                                               276

 

      this, and the timing of this could not have been

 

      better, because we start engaging with our European

 

      and Japanese colleagues on putting decision trees

 

      starting next week, and we needed this leverage to

 

      make the case of where we want to go and hopefully,

 

      bring them along with us, because all of us are in

 

      the same boat right now.

 

                So, I think I really thank the Committee

 

      for the discussion, as well as the strong support

 

      they have given us on this front, and I hope the

 

      pharmaceutical community will really engage and

 

      debate this extensively, so that when we have a

 

      proposal to this committee next time, we actually

 

      can build consensus and move forward.  Otherwise,

 

      this could be a long debate.

 

                The topics for Day 2, today, clearly, I

 

      think show the challenge that we have with respect

 

      to move towards a tactical plan for the type of

 

      questions, the complexity of the questions that you

 

      saw, but in many ways, what we have done is

 

      addressed or attempted to frame a question on

 

      challenges that we have faced for the last 10, 15

 

                                                               277

 

      years.

 

                I am hopeful that bringing a

 

      knowledge-based approach to tackling that problem

 

      might find a solution.  It is not going to be easy,

 

      and clearly, I think we tried to push the agenda in

 

      terms of seeking to tackle a problem like topical

 

      therapeutic equivalence, which will be one of the

 

      most significant challenges.

 

                The challenge will not only be technical,

 

      but also educational, because we will have to

 

      communicate that to the clinicians, the

 

      dermatologists, and the pharmaceutical community in

 

      general.

 

                So, the challenges are not just technical,

 

      but also educational and consensus building across

 

      disciplines to the stakeholders.

 

                With regard to I think highly variable

 

      drugs and the Japanese in many ways have already

 

      made that call.  They are moving.  They have

 

      already applied what we were seeking to apply.

 

                A re-examination of their decision

 

      criteria and putting more rigor to our approach

 

                                                               278

 

      might find a way, not only to seek harmonization

 

      internationally, but also the challenge has always

 

      been that generics are not part of the

 

      International Conference of Harmonization, but

 

      those do things do impact the general decision

 

      trees that we come up with ICH.

 

                So, again, the reason for bringing topics

 

      more focused on generic drug approval was also to

 

      get the generic industry as part and parcel of the

 

      ICH process as much as we could in this discussion.

 

                The key aspect I think I was hoping and

 

      did get the general consensus on is to focus on a

 

      scientific hypothesis driven process, and that is

 

      important because in a regulatory decision

 

      criteria, you need the comfort, as well as the

 

      rigor of a hypothesis testing concept to make

 

      clear-cut decisions.

 

                In many ways, the bioequivalence, although

 

      the goalposts we can argue are arbitrary, and so

 

      forth, but it does give you a sense of decision

 

      which is less arbitrary than it could have been,

 

      and we have been through that transition.

 

                So, in many ways, if you recast the

 

      current requirements that we have on the regulatory

 

      side, like stability testing, bioequivalence, and

 

                                                               279

 

      all of those requirements in the form of a

 

      hypothesis, and your prior knowledge leading to

 

      that hypothesis testing, that provides a way

 

      forward.

 

                I think we will try to construct our

 

      decision trees with that in mind, so as not to add

 

      more burden, but also be relevant in the questions

 

      we ask, and not direct the development program.

 

                So, I think that hopefully, will provide a

 

      common ground to lay out the decision trees.

 

                Our research programs, I think are

 

      critical and thank you for endorsing our request to

 

      have a subcommittee. I am hoping that the working

 

      group or the subcommittee that we form will find

 

      the best practices to lay and create a foundation

 

      for our peer review.

 

                At some point, I think we are initiating

 

      peer review on our review side, and Moheb has

 

      already moved forward in instituting that, that

 

                                                               280

 

      each review of CMC will--not all--but I think

 

      selected reviews will be peer reviewed by their

 

      peers, and at some point I think he will come back

 

      and share with you his thoughts.

 

                Peer review for review is not new.  In

 

      fact, Jerry Collins, when I joined the agency years

 

      ago, we had established a peer review for the

 

      biopharm, and that biopharm day now has really

 

      become a nice model, so I thank Jerry for

 

      initiating some of that thought process, and I

 

      think they are just trying to find the best

 

      practices.

 

                With that, I will stop and thank the

 

      Committee for the valuable information and feedback

 

      that you have provided.

 

                MS. WINKLE:  It looks like every time we

 

      meet, I am recognizing the same two members of the

 

      Advisory Committee, but I do have beautiful plaques

 

      today for Art and Marv for their services as

 

      members of this Advisory Committee.

 

                Obviously, we have enjoyed having them so

 

      much that we haven't let them go away, that we

 

                                                               281

 

      continue to bring them back, because I think they

 

      contribute a whole lot to the conversations and the

 

      discussions we have had.

 

                Anyway, I have some really pretty plaques

 

      for you this time, so I appreciate it.  The last

 

      were just certificates, but these are plaques for

 

      advisors and consultants.

 

                I just have a few things I want to say.  I

 

      have enjoyed listening to the conversations and

 

      discussions over the last two days.  Both of these

 

      topics are topics that I have wanted to discuss for

 

      a long period of time.

 

                For several years now, Ajaz and I have

 

      both been discussing some of the issues over

 

      dissolution, many of the issues over pharmaceutical

 

      equivalence in general, so I was really happy when

 

      we decided to bring these to the Advisory Committee

 

      this time, and to begin to open up our thinking in

 

      these areas.

 

                As Ajaz said earlier when he was talking,

 

      we have actually been learning how to do things for

 

      the last 30 years, and now it is time to apply some

 

                                                               282

 

      of things we have learned, to changing some of the

 

      way we do things.

 

                I think today's and yesterday's

 

      conversations were our good step forward in doing

 

      that.  I started off by talking about the journey

 

      that we were on here and changing the paradigm.  I

 

      think that we took some really significant steps.

 

      They may have seemed small to some people, but they

 

      are very significant to us, I think, in OPS as we

 

      move forward along that pathway.

 

                So, I want to thank you all for your

 

      input.  One of the things, too, I wanted to mention

 

      that up-front, in my opening, I mentioned the fact

 

      that I thought that, in my mind, one of the

 

      initiatives that we have now been working on, the

 

      GMPs for the 21st Century, the Critical Path

 

      Initiative, and the PAT are all leading to a shared

 

      responsibility for product quality.

 

                I think through the shared responsibility,

 

      we need to do more partnering.  We need to partner

 

      with industry, but we also need to partner with

 

      academia, and I think with working through this

 

                                                               283

 

      committee, it is a really good opportunity to

 

      partner and get input from many people who know

 

      things that are going to be beneficial to us as we

 

      do move along our pathway.

 

                So, with that, I want to thank Ajaz.  Ajaz

 

      spent a lot of time putting this together along

 

      with everyone else that gave presentations.  I know

 

      Nozer felt many of them were long, but I think

 

      there were a lot of good points made in these

 

      presentations this time, and I think they were very

 

      worthwhile in helping us get a better understanding

 

      of some of the issues that we had to tackle.

 

                Also, I want to thank Bob King, who herds

 

      us all through this.  It is just like herding cats,

 

      believe me, and he really deserves a lot of thanks

 

      for that.

 

                I appreciate, too, your input on the

 

      subcommittee. I think this is very going to be very

 

      valuable to OPS to take a look at the two research

 

      programs and see how we can better coordinate and

 

      set priorities, et cetera, so I appreciate that.

 

                With that, I will close and turn it back

 

                                                               284

 

      over to Charlie.

 

                DR. COONEY:  Thank you.  My apologies.

 

                Art?

 

                DR. KIBBE:  Listening to the summary made

 

      me think of one thing that I had written down and

 

      forgot to say.  I think it would be well if the

 

      agency could assure the industry that when they

 

      provide drug product development information, that

 

      the reviewer who reviews it will have a working

 

      knowledge of drug product development and some

 

      hands-on experience with the equipment and the

 

      materials that are being used to do drug product

 

      development, so that the review is worthwhile on

 

      both ends of the thing.

 

                Second, thanks for the plaque.  It has

 

      really been fun irritating Ajaz all these years,

 

      and I continue to look forward to having an

 

      opportunity to continue to do that.

 

                DR. COONEY:  Thank you all very much and I

 

      think I can now close the meeting without creating

 

      yet another faux pas.  Thank you.

 

                [Whereupon, at 3:30 p.m., the meeting was

 

                                                               285

 

      concluded.]

 

                                 - - -