1

 

                DEPARTMENT OF HEALTH AND HUMAN SERVICES

 

                      FOOD AND DRUG ADMINISTRATION

 

                CENTER FOR DRUG EVALUATION AND RESEARCH

 

 

 

               PEDIATRIC ONCOLOGY SUBCOMMITTEE COMMITTEE

 

               OF THE ONCOLOGIC DRUGS ADVISORY COMMITTEE

 

 

 

 

 

                       Wednesday, March, 17, 2004

 

                               8:00 a.m.

 

 

 

                           5630 Fishers Lane

                      First Floor Conference Room

                          Rockville, Maryland

 

                                                                 2

                              PARTICIPANTS

 

      ONCOLOGIC DRUGS ADVISORY COMMITTEE:

 

         Donna Przepiorka, MD., Ph.D., Chair, ODAC

         Johanna Clifford, M.S., RN, BSN

 

         Pamela J. Haylock, RN,

           Consumer Representative, ODAC

 

      CONSULTANTS (VOTING):

 

         Victor Santana, M.D., Chair

         Peter Adamson, M.D.

         Alice Ettinger, M.S., RN

         Peter Houghton, Ph.D.

         Eric Kodish, M.D.

         C. Patrick Reynolds, M.D., Ph.D.

         Susan Weiner, Ph.D.

         Ruth Hoffman, Patient Representative

         Barry Anderson, M.D., Ph.D.

         Lee J. Helman, M.D.

         Malcolm Smith, M.D., Ph.D.

         Paul Meltzer, M.D.

         Chand Khanna, DVM, Ph.D., DACVIM

         Kenneth Hastings, Ph.D.

      ACTING INDUSTRY REPRESENTATIVE (NON-VOTING):

 

         Antonio Grillo-Lopez, M.D.,

 

      FDA STAFF:

 

         Susan Ellenberg, Ph.D.

         Steven Hirschfeld, M.D., Ph.D.

         Ramzi Dagher, M.D.

         Richard Pazdur, M.D.

         Patricia Keegan, M.D.

         Pat Dinndorf, M.D.

         Grant Williams, M.D.

 

                                                                 3

 

                            C O N T E N T S

 

                                                              PAGE

 

      Call to Order, Victor Santana, M.D., Chair                 5

 

      Introductions                                              5

 

      Conflict of Interest Statement,

         Johanna Clifford, M.S., RN, BSN                         7

 

      Safety Monitoring in Clinical Studies Enrolling

      Children with Cancer:

 

      Opening Remarks, Richard Pazdur, Director,

         Division of Oncology Drug Products, FDA                10

 

      Introduction of Issues and Agenda,

         Steven Hirschfeld, M.D., Ph.D., Office

         of Cellular and Gene Therapy, FDA                      11

 

      Protecting Children in Cancer Research: What Really

         Matters, Eric Kodish, M.D., Director, Rainbow

         Center for Pediatric Ethics                            23

 

      Legal Responsibilities for HHS Supported Studies,

         Michael Carome, M.D., Office for Human Research

         Protection, HHS                                        49

 

      Legal Responsibilities for Studies with FDA

         Regulated Products, Steven Hirschfeld, M.D.,

         Ph.D., Office of Cellular and Gene Therapy,

         CBER, FDA                                              61

 

      Enrollment and Monitoring Procedures for NCI Funded

         Studies, Barry Anderson, M.D., Ph.D., Cancer

         Treatment Evaluation Program, NCI                      70

 

      Open Public Hearing:

         Wayne Rakoff, Johnson & Johnson                        87

 

      Monitoring Procedures in a Private

         Children's Hospital,

         Victor Santana, M.D., St. Jude

           Children's Hospital                                  88

 

      Committee Discussion                                     116

 

      Questions for Discussion                                 154

 

                                                                 4

 

                      C O N T E N T S (Continued)

 

                                                              PAGE

      Use of Nonclinical Data to Complement Clinical Data

      for Pediatric Oncology:

 

      What are Microarrays and How Can They Help Us

         with Clinical Studies in Pediatric Oncology,

         Paul Meltzer, National Human Genome Research

         Institute, NIH                                        198

 

      Advantages and Limitations of Cell Culture Models

         in Pediatric Drug Developments,

         Peter Adamson, M.D., Children's Hospital

         of Philadelphia                                       210

 

      Human Cell-Animal Xenografts: The Current Status,

         Potential and Limits of Informing Us About

         Clinical Studies, Peter Houghton, Ph.D., St.

         Jude Children's Research Hospital                     229

 

      An Integrated and Comparative Approach to

         Preclinical/Clinical Drug Development,

         Chand Khanna, DVM, Ph.D.,

         Tumor and Metastasis Biology Section, NIH             248

 

      What Can be Learned About Safety,

         Kenneth Hastings, Ph.D., CDER, FDA                    263

 

      Assessing Anti-Tumor Activity in Nonclinical Models

         of Childhood Cancer, Malcolm Smith, M.D., Ph.D.,

         Treatment Evaluation Program, National Cancer

         Institutes, NIH                                       280

 

      Committee Discussion                                     294

 

      Questions for Discussion                                 306

 

                                                                 5

 

  1                      P R O C E E D I N G S

 

  2                          Call to Order

 

  3             DR. SANTANA:  Good morning to everyone.  I

 

  4   know Dr. Kodish is on the line so good morning to

 

  5   you too, Eric.  I hope you can hear us well.

 

  6             DR. KODISH:  Good morning, Victor.

 

  7             DR. SANTANA:  This is a meeting of the

 

  8   Pediatric Oncology Subcommittee of the Oncology

 

  9   Drugs Advisory Committee and we are here today to

 

 10   advise the agency on two issues.  In the morning we

 

 11   will deal with the issue of safety monitoring in

 

 12   clinical studies enrolling pediatric oncology

 

 13   patients.  Then, in the afternoon we will address

 

 14   issues related to the use of nonclinical data to

 

 15   complement clinical data for proposed pediatric

 

 16   oncology studies.  So, we have quite a busy agenda

 

 17   and I think we will go ahead and get started with

 

 18   the introductions, and I am feeling so sorry for

 

 19   Dr. Anderson who is sitting all by himself over

 

 20   there, but we will go ahead and get started with

 

 21   him and then move around.

 

 22                          Introductions

 

 23             DR. ANDERSON:  Barry Anderson, from NCI

 

 24   CTEP.

 

 25             DR. GRILLO-LOPEZ:  Antonio Grillo-Lopez,

 

                                                                 6

 

  1   Neoplastic and Autoimmune Diseases Disorders

 

  2   Research Institute.

 

  3             DR. WEINER:  I am Susan Weiner, from The

 

  4   Children's Cause, a patient advocate.

 

  5             MS. HOFFMAN:  Ruth Hoffman, patient

 

  6   advocate.

 

  7             DR. PRZEPIORKA:  Donna Przepiorka,

 

  8   University of Tennessee, Memphis.

 

  9             MS. CLIFFORD:  Johanna Clifford, executive

 

 10   secretary to this meeting.

 

 11             DR. SANTANA:  Victor Santana, pediatric

 

 12   oncologist at St. Jude Children's Research

 

 13   Hospital, Memphis, Tennessee.

 

 14             DR. REYNOLDS:  Dr. Reynolds, Children's

 

 15   Hospital of Los Angeles.

 

 16             MS. ETTINGER:  Alice Ettinger, pediatric

 

 17   nurse practitioner, St. Peter's University Hospital

 

 18   in New Jersey.

 

 19             DR. PAZDUR:  This is Susan Ellenberg, who

 

 20   has laryngitis.  She is a statistician.  I am

 

 21   Richard Pazdur.

 

 22             DR. HIRSCHFELD:  Steven Hirschfeld, FDA.

 

 23             DR. DINNDORF:  Patricia Dinndorf, FDA.

 

 24             DR. DAGHER:  Ramzi Dagher, FDA.

 

 25             DR. SANTANA:  Eric, will you go ahead and

 

                                                                 7

 

  1   announce your name and affiliation for the record?

 

  2             DR. KODISH:  I am Eric Kodish, from

 

  3   Cleveland, Ohio, Rainbow Babies & Children's

 

  4   Hospital.

 

  5             DR. SANTANA:  Thank you, Eric.  With that,

 

  6   we will go ahead and have Ms. Clifford read us the

 

  7   conflict of interest statement.

 

  8                  Conflict of Interest Statement

 

  9             MS. CLIFFORD:  Thank you.  The following

 

 10   announcement addresses conflict of interest issues

 

 11   associated with this meeting and is made a part of

 

 12   the record to preclude even the appearance of such

 

 13   at this meeting.

 

 14             Based on the agenda, it has been

 

 15   determined that the topics of today's meeting are

 

 16   issues of broad applicability and there are no

 

 17   products being approved at this meeting.  Unlike

 

 18   issues before a committee in which a particular

 

 19   product is discussed, issues of broader

 

 20   applicability involve many industrial sponsors and

 

 21   academic institutions.

 

 22             All special government employees have been

 

 23   screened for their financial interests as they may

 

 24   apply to the general topics at hand.  To determine

 

 25   if any conflict of interest existed, the agency has

 

                                                                 8

 

  1   reviewed the agenda and all relevant financial

 

  2   interests reported by the meeting participants.

 

  3   The Food and Drug Administration has granted

 

  4   general matters waivers to the special government

 

  5   employees participating in this meeting who require

 

  6   a waiver under Title 18, United States Code,

 

  7   Section 208.

 

  8             A copy of the waiver statements may be

 

  9   obtained by submitting a written request to the

 

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

 

 11   of the Parklawn Building.

 

 12             Because general topics impact so many

 

 13   entities, it is not prudent to recite all potential

 

 14   conflicts of interest as they apply to each member

 

 15   and consultant and guest speaker.  FDA acknowledges

 

 16   that there may be potential conflicts of interest

 

 17   but, because of the general nature of the

 

 18   discussion before the committee, these potential

 

 19   conflicts are mitigated.

 

 20             With respect to FDA's invited industry

 

 21   representative, we would like to disclose that Dr.

 

 22   Antonio Grillo-Lopez is participating in this

 

 23   meeting as an acting industry representative,

 

 24   acting on behalf of regulated industry.  Dr.

 

 25   Grillo-Lopez is employed by Neoplastic and

 

                                                                 9

 

  1   Autoimmune Diseases Research.

 

  2             In the event that the discussions involve

 

  3   any other products or firms not already on the

 

  4   agenda for which FDA participants have a financial

 

  5   interest, the participants' involvement and their

 

  6   exclusion will be noted for the record.

 

  7             With respect to all other participants, we

 

  8   ask in the interest of fairness that they address

 

  9   any current or previous financial involvement with

 

 10   any firm whose product they may with to comment

 

 11   upon.  Thank you.

 

 12             DR. SANTANA:  Thanks, Johanna.  Anybody

 

 13   else sitting at the table that wants to disclose

 

 14   anything publicly?  No?  Dr. Adamson just joined

 

 15   the group.  Do you want to introduce yourself,

 

 16   Peter, please?

 

 17             DR. ADAMSON:  Peter Adamson, from

 

 18   Children's Hospital of Philadelphia.

 

 19             DR. SANTANA:  Thanks, Peter.  Peter, do

 

 20   you want to introduce yourself?

 

 21             DR. HOUGHTON:  Peter Houghton, St. Jude

 

 22   Children's Research Hospital.

 

 23             DR. SANTANA:  With that, I will pass it

 

 24   over to Dr. Pazdur for his opening remarks.

 

 25                         Opening Remarks

 

                                                                10

 

  1             DR. PAZDUR:  Well, I would like to

 

  2   disclose something publicly, my disappointment with

 

  3   Victor and Johanna for not mentioning this but the

 

  4   disclosure is happy St. Patrick's Day.

 

  5             [Laughter]

 

  6             As you can see, we in the government have

 

  7   provided you with green folders for the day and,

 

  8   obviously, I am dressed in green but I would like

 

  9   to remind you Pazdur is not an Irish name.  The

 

 10   other thing I would like to just emphasize is that

 

 11   Donna and I, as compatriots from Chicago's Polish

 

 12   community, would like to emphasize that St.

 

 13   Patrick's Day is just a warm-up for St. Joseph's

 

 14   Day.  Okay?

 

 15             [Laughter]

 

 16             DR. SANTANA:  Which is Friday, March 19th.

 

 17             DR. PAZDUR:  Thanks for pointing that out.

 

 18             In all seriousness, I would like to go

 

 19   back to why we are here today, and that is for the

 

 20   subcommittee to discuss two important areas today,

 

 21   one in the morning discussing safety monitoring in

 

 22   clinical studies enrolling children with cancer and

 

 23   then, in the afternoon, discussing nonclinical data

 

 24   to complement clinical data for pediatric oncology.

 

 25             We look at these as very important

 

                                                                11

 

  1   thematic discussions to have.  How these areas

 

  2   impact on oncology drug development I think is very

 

  3   important.  One thing that I would ask the

 

  4   committee to do specifically is to concentrate

 

  5   really on the pediatric aspect of these.  I know

 

  6   that these areas have some tentacles to adult

 

  7   oncology and to other areas of oncology but I would

 

  8   like to remind you that the purpose of this

 

  9   subcommittee is to focus on the pediatric

 

 10   specificity of these issues and special

 

 11   considerations of these broad issues in pediatric

 

 12   oncology.

 

 13             I would like to thank everyone for being

 

 14   here.  I asked Steve what number meeting this is

 

 15   and we think it is the eighth.  We may be wrong but

 

 16   we are happy that the committee is meeting on a

 

 17   regular basis.  We intend to have the committee

 

 18   meet on a regular basis here and to continue this

 

 19   dialogue with the community.  So, Steve, I will

 

 20   turn it over to you.

 

 21                Introduction of Issues and Agenda

 

 22             DR. HIRSCHFELD:  Thank you.  It is

 

 23   customary at the end of remarks to give the

 

 24   acknowledgments but I wanted to give two

 

 25   acknowledgments initially.  The first one is to

 

                                                                12

 

  1   someone who is in the room right now and I am

 

  2   looking at her, and that is Johanna Clifford who

 

  3   has done I think a marvelous job in helping to

 

  4   organize this meeting, and we have had a number of

 

  5   challenges to overcome along the way, so many

 

  6   challenges that for a period of time we thought we

 

  7   were working under a curse, but Johanna has been

 

  8   steadfast, good humored, competent, rapid in her

 

  9   responses and has been I think a driving force in

 

 10   terms of having the meeting occur as it is and as

 

 11   well organized as it is today.  So, thank you,

 

 12   Johanna.

 

 13             I would also like to acknowledge someone

 

 14   who is in this room, although not physically, but

 

 15   someone who has had enormous influence on our

 

 16   thinking and on our policies toward patients

 

 17   enrollment in studies and in particular children

 

 18   enrolling in studies, and that is Bonnie Lee who

 

 19   has been with the FDA for many years and was

 

 20   associated with the initial hearings of the

 

 21   committee, which was mandated by Congress in the

 

 22   1970s, to examine the role of children in clinical

 

 23   research.  Bonnie has been a particular guide and

 

 24   inspiration for me and also a source of information

 

 25   and direction, which I think has been an asset not

 

                                                                13

 

  1   only to the agency but to the country and to all

 

  2   patients.  And, I wanted to dedicate the discussion

 

  3   this morning in her honor.  So, thank you, Bonnie.

 

  4             As Dr. Pazdur pointed out, we are going to

 

  5   be discussing the themes of safety and

 

  6   extrapolation.  Clinical research, which we have

 

  7   discussed in some detail in this forum over several

 

  8   of the meetings, has been recorded for at least

 

  9   2,400 years.  Children were often the first

 

 10   patients for new procedures and interventions.

 

 11   Part of this evolved from the concept that children

 

 12   were the property of parents so it was rather easy

 

 13   for parents to donate their children for whatever

 

 14   questions might be asked.  But along the way there

 

 15   were some founding principles because,

 

 16   unfortunately, children have also been the victims

 

 17   of clinical research.

 

 18             The founding principles of modern Food and

 

 19   Drug Administration regulation were, in large part,

 

 20   established for the purpose of protecting children

 

 21   and, yet, pediatric therapeutic development has

 

 22   never been as thorough and robust as adult

 

 23   therapeutic development, and most of the people in

 

 24   this room have been part of that process and

 

 25   witness to these inequities.  Many therapies are

 

                                                                14

 

  1   administered to children without adequate studies

 

  2   and, furthermore, many therapies are not made

 

  3   available for pediatric study until after adult

 

  4   marketing studies are completed and this is

 

  5   particularly true in oncology.  So, we have been

 

  6   working to overcome some of these barriers and

 

  7   challenges.  And, the challenges are to assemble

 

  8   sufficient data to establish efficacy and safety in

 

  9   the relevant population.  The relevant population

 

 10   may be sufficiently rare that confirmatory studies

 

 11   are not feasible, which is particularly the case

 

 12   for many of the childhood malignancies.

 

 13             There are concerns regarding the

 

 14   implications of adverse events in children and this

 

 15   has been a barrier to the further clinical

 

 16   development of some products because of these

 

 17   concerns.  It is also important that there is the

 

 18   establishment and maintenance of a framework that

 

 19   would support systematic clinical investigations

 

 20   for the relevant population.  This has been the

 

 21   case historically in pediatric oncology but that

 

 22   framework has always been challenged and is always

 

 23   competing with other priorities.  So, it is

 

 24   incumbent on us to make sure that that pediatric

 

 25   research framework has the best resources, and the

 

                                                                15

 

  1   best advice, and the best support, and the best

 

  2   regulatory environment to do its job.

 

  3             The particular issues regarding the safety

 

  4   monitoring in pediatric oncology clinical

 

  5   investigations are an acknowledgment that children

 

  6   require special protections.  Yet, on the other

 

  7   hand, there is also an acknowledgment that risk

 

  8   tolerance is higher in oncology therapeutics than

 

  9   in other therapeutic areas.  This sets up a

 

 10   potential tension.  Furthermore, there are no

 

 11   detailed consensus standards on study monitoring

 

 12   despite numerous international documents describing

 

 13   what could be termed good clinical practice.  We

 

 14   will examine those in some detail during the course

 

 15   of the morning.  So, the charge to the committee is

 

 16   to suggest ways to incorporate the fundamental

 

 17   ethical and scientific principles in protecting

 

 18   patients enrolled in clinical studies for pediatric

 

 19   malignancies while providing clear guidance and

 

 20   minimizing the resource burden.

 

 21             We have a series of questions directed

 

 22   toward the committee to help focus the discussion.

 

 23   These are questions which are meant to stimulate

 

 24   what we hope will be an informative exchange and do

 

 25   not have a yes/no or a definitive answer.

 

                                                                16

 

  1             The first questions revolves around the

 

  2   principles, what are the principles that should be

 

  3   addressed in safety monitoring of clinical studies

 

  4   that enroll children with cancer?  Dr. Kodish is

 

  5   going to provide us with some background on that

 

  6   particular topic.  If the principles are adequately

 

  7   stated in existing documents. statutes or

 

  8   regulations, please identify the relevant documents

 

  9   and sections.

 

 10             The second set of questions deals with the

 

 11   practice.  Recognizing that particular populations,

 

 12   disease settings and products may have specific

 

 13   requirements, what general parameters should be

 

 14   monitored for safety in all clinical studies?  Or,

 

 15   to rephrase that, what should the default position

 

 16   be for safety monitoring?

 

 17             Based on the response to the previous

 

 18   question, how often should these parameters be

 

 19   monitored?  Again, just giving a framework or

 

 20   guidelines.

 

 21             Based on the responses to the previous

 

 22   questions, who should do the monitoring?  Is it

 

 23   adequate to have the personnel involved in the

 

 24   study be responsible for safety monitoring?  When

 

 25   we discuss this in detail we may parse this out

 

                                                                17

 

  1   into the type of study, whether it is early

 

  2   development or later development or the type of

 

  3   disease or other risk factors.

 

  4             What circumstances would benefit from a

 

  5   data monitoring committee?  And, are there

 

  6   additional recommendations for safety monitoring?

 

  7             The afternoon will be devoted to a

 

  8   question which can be traced back to the principle

 

  9   of extrapolation.  Extrapolation has been a topic

 

 10   of interest within the Food and Drug Administration

 

 11   for many years.  In recent years there has been an

 

 12   FDA working group on pediatric extrapolation that

 

 13   has identified four domains that may provide a

 

 14   basis for extrapolation of adult data to the

 

 15   pediatric population.  These are nonclinical data,

 

 16   pathophysiology, natural history of the disease or

 

 17   condition, and response to therapy.

 

 18             When our group, noted at the bottom of the

 

 19   slide and some of the members are present here in

 

 20   the audience, asked ourselves the question how can

 

 21   we use nonclinical data to inform us about

 

 22   pediatric clinical studies, and in particular

 

 23   pediatric studies in clinical oncology, we realized

 

 24   we needed further background and further discussion

 

 25   before we could have an informed approach to it.

 

                                                                18

 

  1             We recognize that the absence of

 

  2   predictive or explanatory nonclinical models in

 

  3   pediatric oncology is today's status quo.  We know

 

  4   that safety prediction based on animal studies is

 

  5   estimated at approximately 65-70 percent for

 

  6   cytotoxic compounds and it is unknown for other

 

  7   classes of compounds, particularly the new biologic

 

  8   therapies, gene therapies, immunotherapy, and

 

  9   cellular-based therapies.  Efficacy prediction is

 

 10   unknown but low at best.  The findings in clinical

 

 11   studies, particularly negative studies, often

 

 12   remain unexplained.

 

 13             Therefore, further clinical studies that

 

 14   entail resources and risks are undertaken to

 

 15   further the field, and we are posing the paradigm

 

 16   is there a mechanism by which we can use

 

 17   nonclinical data to inform us and improve the

 

 18   clinical research in pediatric oncology.  There are

 

 19   potential advantages of using the nonclinical data:

 

 20   a lesser resource burden; the ability to answer

 

 21   questions not amenable to available clinical

 

 22   techniques.  There might be ethical or, in fact,

 

 23   legal considerations involved too; possibly a

 

 24   faster time frame to generate data; a dynamic

 

 25   interaction between clinical and nonclinical

 

                                                                19

 

  1   findings that can enhance understanding and

 

  2   confidence in results.  When we only have a

 

  3   sufficient population to do one definitive study,

 

  4   and that study takes three to five years and it is

 

  5   not feasible to do a confirmatory study, having

 

  6   confidence in those results is critical.  The

 

  7   avoidance of non-informative and minimization of

 

  8   negative outcome studies could be another outgrowth

 

  9   and an opportunity for new study designs.

 

 10             So, the charge to the committee for this

 

 11   afternoon is to provide advice on what types of

 

 12   nonclinical data are considered informative to

 

 13   complement or supplement clinical results.  What

 

 14   should the characteristics or properties of

 

 15   nonclinical models and data be to effectively add

 

 16   to the clinical results?

 

 17             If there are no satisfactory models that

 

 18   exist currently, and we will hear some discussion

 

 19   on approaches, what characteristics should a

 

 20   nonclinical model have to confirm, extend or

 

 21   substitute for clinical results?

 

 22             Lastly, is there a set of postulates that

 

 23   can be identified, or should a set be developed to

 

 24   help us make the transition for data extrapolation?

 

 25   So, the questions we are asking are what types of

 

                                                                20

 

  1   questions that are of potential clinical relevance

 

  2   but are not feasible or acceptable to answer in a

 

  3   clinical study could be addressed by nonclinical

 

  4   studies.

 

  5             Examples may include the need for repeated

 

  6   tissue sampling, always a contentious issue,

 

  7   particularly in children; the assessment of

 

  8   long-term effects of treatment; effects on

 

  9   reproduction; access to critical anatomic

 

 10   structures, and this is a consideration again

 

 11   particularly for some of the pediatric brain

 

 12   tumors; exposure to toxic reagents; evaluation of

 

 13   non-monitorable or irreversible toxicities;

 

 14   identification of biomarkers for clinical

 

 15   monitoring; and many others which I am sure will

 

 16   come up when we have our learned and motivated

 

 17   panel discuss the issue.

 

 18             What type of evidence and data would be

 

 19   recommended in each of the following domains to

 

 20   allow extrapolation from nonclinical data and be

 

 21   informative for a clinical condition?  There are

 

 22   listed here a few but there may be others.  These

 

 23   include, but are not limited to pharmacology and

 

 24   pharmacokinetics, safety, efficacy, behavior,

 

 25   long-term effects, developmental aspects and others

 

                                                                21

 

  1   which I am sure will come up.

 

  2             Are there additional recommendations for

 

  3   the effective use of nonclinical data?  For

 

  4   example, will open literature reports be generally

 

  5   acceptable?  Is documentation of compliance with

 

  6   Good Laboratory Practice necessary to evaluate

 

  7   animal data?  Should nonclinical data be submitted

 

  8   as an independent report with a presentation of

 

  9   primary data sufficient for verification and

 

 10   review?  These are all practical questions and we

 

 11   are looking for specific advice.

 

 12             So, with this charge and these questions

 

 13   before you, I would like to thank all the committee

 

 14   members and our speakers and guests, and everyone

 

 15   who has shown an interest here for participating in

 

 16   this discussion, and I will turn now the further

 

 17   presentation over to Dr. Eric Kodish, who will

 

 18   discuss the fundamental principles involved in

 

 19   clinical research and some of the issues of

 

 20   enrolling children.

 

 21             Dr. Santana, I think perhaps before we

 

 22   have Dr. Kodish speak--we have some more members of

 

 23   the panel that should be introduced.

 

 24             DR. SANTANA:  Yes.  Anybody that joined us

 

 25   a little bit late, could you please identify

 

                                                                22

 

  1   yourself into the microphone by name and

 

  2   affiliation, and any potential conflicts that may

 

  3   have arisen since we started?

 

  4             MS. HAYLOCK:  I am Pam Haylock.  I am an

 

  5   oncology nurse and I am at the University of Texas

 

  6   Medical Branch, in Galveston.

 

  7             DR. SMITH:  I am Malcolm Smith, pediatric

 

  8   oncologist at the Cancer Therapy Evaluation

 

  9   Program, NCI.

 

 10             DR. SANTANA:  Dr. Grillo, you had your

 

 11   hand up?

 

 12             DR. GRILLO-LOPEZ:  Yes, a point of

 

 13   clarification that I would like to propose to Dr.

 

 14   Hirschfeld.  On his first slide on the charge to

 

 15   the committee, which addresses the morning session,

 

 16   you used the phrase "providing clear guidance and

 

 17   minimizing the resource burden" which clearly

 

 18   applies to human resources and financial resources

 

 19   but perhaps doesn't quite stress time.  I would

 

 20   suggest that part of your charge to the committee

 

 21   should be that whatever recommendations we propose,

 

 22   and however the FDA understands and decides to

 

 23   apply those recommendations, should not affect the

 

 24   time lines for cancer drug development which today

 

 25   are already intolerably long, and we should be

 

                                                                23

 

  1   concerned that the cancer patient in general should

 

  2   not be subject to those too long time lines and

 

  3   that anything we do should, in fact, try to reduce

 

  4   the time lines for approval of new therapies.

 

  5             DR. HIRSCHFELD:  Thank you for your

 

  6   comments, Dr. Grillo-Lopez.  I think you touched on

 

  7   one of the themes which is implied.  I personally

 

  8   have always incorporated in the concept resource of

 

  9   time because time is, in fact, probably the most

 

 10   precious resource and, if one looks at biology as a

 

 11   broad spectrum, time is something which evolution

 

 12   and biologic processes look to, to conserve in many

 

 13   ways too.  So, I thank you for calling attention to

 

 14   the issue of time, and it is incorporated in that

 

 15   specific charge.

 

 16             DR. SANTANA:  One of the philosophic

 

 17   principles of stewardship is that it involves time,

 

 18   people and money resources.  So, I think those are

 

 19   all encompassed in your comments.

 

 20             With that, Eric, are you on line now?  Can

 

 21   we proceed with you?

 

 22             DR. KODISH:  I am on line, Victor.

 

 23             DR. SANTANA:  Good.  Go ahead, Eric.

 

 24             Protecting Children in Cancer Research:

 

 25                       What Really Matters

 

                                                                24

 

  1             DR. KODISH:  Good morning.  It is good to

 

  2   be with you virtually, if not physically.  I

 

  3   apologize for the inability to get to Washington.

 

  4   We have, hopefully, completed our last big

 

  5   snowstorm of the winter in Cleveland.

 

  6             I am going to be speaking this morning

 

  7   over the telephone and looking at a Webcast of the

 

  8   slides and this is a work in progress so, please,

 

  9   interrupt me if it is not going well and I will

 

 10   switch to my Power Point presentation. I am looking

 

 11   at the Webcast now and I don't see my Power Point

 

 12   slides yet.  What I plan to do is ask Johanna to

 

 13   put on the next slide before I move through them.

 

 14   So, let's give it a moment for me to see the first

 

 15   slide.

 

 16             I can introduce the talk by saying that I

 

 17   have always thought I had a face for radio and this

 

 18   is an example of that perhaps--

 

 19             [Laughter]

 

 20              I see my first slide.  the title of this

 

 21   presentation is "Protecting Children in Cancer

 

 22   Research: What Really Matters."

 

 23             Can I ask that we have the next slide,

 

 24   please?

 

 25             MS. CLIFFORD:  You know what, Dr. Kodish,

 

                                                                25

 

  1   if you just want to move on through your

 

  2   presentation--

 

  3             DR. KODISH:  I have it now.  Should I go

 

  4   to the Power Point instead?

 

  5             MS. CLIFFORD:  Yes, that would be great.

 

  6             DR. KODISH:  All right, the Webcast didn't

 

  7   work well and I will look forward to joining you on

 

  8   the Webcast after I have done my talk.

 

  9             MS. CLIFFORD:  Okay, there just seems to

 

 10   be a delay.

 

 11             DR. KODISH:  I figured that might happen.

 

 12   The Belmont report I think articulates the key

 

 13   principles of research involving human subjects.

 

 14   My purpose today is to respond to the charge that

 

 15   has been given to the committee and to paint in

 

 16   broad strokes what the key principles are for

 

 17   protection of children involved in cancer research.

 

 18   I think it starts with the Belmont report and the

 

 19   three key principles that are articulated there are

 

 20   beneficence, respect for persons and justice.

 

 21             The next slide, please.  This slide shows

 

 22   a concept of principles that move into practice.  I

 

 23   thought it was quite appropriate that the charge

 

 24   for the first half of the meeting talked about both

 

 25   principles and practice.  I view the regulations

 

                                                                26

 

  1   and their interpretation as a conduit, as a

 

  2   mechanism by which we move from principles to

 

  3   practice.  I want to emphasize the word

 

  4   "interpretation" here.  I think that the current

 

  5   set of regulations is subject to wide

 

  6   interpretation, as has been pointed out over and

 

  7   over again in the literature.  I don't view this as

 

  8   a negative.  I think that it allows for thoughtful

 

  9   IRBs, investigators, parents and others involved in

 

 10   the research process to move from principles to

 

 11   practice in an appropriate manner, and that

 

 12   interpretation is really the key step.

 

 13             The next slide, please.  This slide should

 

 14   show a triangle which points out that we are

 

 15   talking today about pediatric research ethics and

 

 16   that this is a more complicated system because of

 

 17   the involvement of a child.  The geometry of

 

 18   pediatric research ethics involves parents, on your

 

 19   lower left; the investigator, on your lower right;

 

 20   and the child at the top of the triangle.  If we

 

 21   keep the best interests of the child in mind at all

 

 22   points, I think we will be responding to perhaps

 

 23   the most fundamental issue in research involving

 

 24   children.

 

 25             The next slide, please.  This slide shows

 

                                                                27

 

  1   a recapitulation of the Belmont principles with an

 

  2   emphasis on beneficence in pediatric ethics.

 

  3   Respect for persons and justice remain important in

 

  4   pediatric ethics but it is my feeling that there is

 

  5   a special place for beneficence when we are talking

 

  6   about children, whether it is research involving

 

  7   children or in clinical ethics regarding children.

 

  8   In fact, more broadly in social policy regarding

 

  9   children it is important to remember that children

 

 10   are not able to vote; don't have economic

 

 11   resources; and we owe an advocacy role I think on

 

 12   behalf of children.  It is very important and, to

 

 13   me, prioritizes that beneficence as a concept for

 

 14   pediatric ethics.

 

 15             Can I have the next slide, please?  The

 

 16   principles of medical ethics then are different for

 

 17   children compared with adults.  I would say that

 

 18   respect for persons, for good or for bad, has

 

 19   become the dominant principle for adult ethics and

 

 20   this is seen in research ethics where there is a

 

 21   tremendous emphasis on informed consent, and this

 

 22   is out of the derivative concept of autonomy which

 

 23   comes from that principle of respect for persons.

 

 24   By contrast, as I said, I think the best interest

 

 25   of children has to dominate pediatric ethics and

 

                                                                28

 

  1   justifies an population that takes beneficence as

 

  2   the most important principles.

 

  3             I don't want you to move slides back but,

 

  4   if you recall a few slides ago, the slide that

 

  5   shows moving principles into practice, I think

 

  6   beneficence has to be the principle that drives our

 

  7   interpretation of the regulations and our actual

 

  8   practices.

 

  9             The next slide, please.  This slide

 

 10   dissects out some text from the Belmont report.

 

 11   The document itself talks about beneficence as an

 

 12   obligation with two general rules.  These are very

 

 13   interesting.  It had been sometime since I have

 

 14   looked at them and in preparing for this

 

 15   presentation I found the two general rules cited by

 

 16   Belmont are do not harm and, secondly, maximize

 

 17   possible benefits and minimize possible harms.

 

 18             On the face of it, these two general rules

 

 19   can be read as conflicting with one another.  That

 

 20   is, the charge do not harm is an absolute standard,

 

 21   whereas in the second rule of minimizing possible

 

 22   harms and maximizing possible benefits it is a

 

 23   relative standard and it calls for a weighing of

 

 24   benefit against harm.  Again, to put interpretation

 

 25   into play, I think it is the second rule that is

 

                                                                29

 

  1   most appropriate for pediatric oncology studies.

 

  2   That is to say, if one is talking about research

 

  3   involving healthy children with no prospect of

 

  4   benefit to that child, the first rule might be more

 

  5   appropriate to apply, do not harm, period.  But we

 

  6   are talking about a balance in pediatric oncology

 

  7   and I think the second general rule is more

 

  8   appropriate.

 

  9             Can I have the next slide, please?  If we

 

 10   are on the same page, this slide should continue to

 

 11   cite the Belmont report which says that beneficence

 

 12   is not always so unambiguous and goes on to say

 

 13   that prohibiting research that presents more than

 

 14   minimal risk without the immediate prospect of

 

 15   direct benefit to the children involved limits

 

 16   potential for great benefit to children in the

 

 17   future.

 

 18             This became, in some sense, the foundation

 

 19   for the different categories of research in subpart

 

 20   D that IRBs are able to approve and points out the

 

 21   key ethical dilemma, as far as I am concerned,

 

 22   which has to do with how we weigh benefits or which

 

 23   benefits count when we are weighing risk and

 

 24   benefit.

 

 25             The next slide, please.  The subtitle of

 

                                                                30

 

  1   my talk today is "What Really Matters" and as I

 

  2   thought about a way of presenting this I decided

 

  3   that it could be divided in three phases, what

 

  4   matters before a clinical trial begins; what

 

  5   matters during the conduct of the trial; and what

 

  6   matters after a trial has closed.

 

  7             One of the members of the panel pointed

 

  8   out the importance of time prior to the beginning

 

  9   of my talk, and I guess this is another way of

 

 10   looking at time as a divider for where the

 

 11   different ethical obligations come in.

 

 12             Speaking of time, I wanted to get some

 

 13   validation from Johanna.  Is the timing going

 

 14   better now with the slides?

 

 15             MS. CLIFFORD:  It is fine, Dr. Kodish.

 

 16             DR. KODISH:  Going fine?  Great!  So, I

 

 17   would like to now talk about what matters before a

 

 18   trial begins and I could think of at least three

 

 19   important issues.  The first is that it be

 

 20   significant science.  Again, interpretation is a

 

 21   key here.  My view of significant science is that

 

 22   it has the potential to help children with cancer.

 

 23   I think it is important that I am very specific

 

 24   about that.  I think that if there are going to be

 

 25   exposures of risk to children with cancer the

 

                                                                31

 

  1   potential to help children with cystic fibrosis,

 

  2   for example, may not be considered significant

 

  3   science by this test.  The potential to help adults

 

  4   with Alzheimer's disease may not be significant

 

  5   science by this test.

 

  6             I think that we need to be cognizant of

 

  7   the fact that research involving children with

 

  8   cancer needs to resound back to help children with

 

  9   cancer and that one should look for other avenues

 

 10   to study other important diseases.  It is difficult

 

 11   to think of children with cancer as a resource, but

 

 12   I think in some sense this really forces us to do

 

 13   that and, by limiting the risk of exposure to

 

 14   children to that which will come back to help

 

 15   children--and I know that scientifically it is

 

 16   often very difficult to predict in which direction

 

 17   the work will go and how the results will, in fact,

 

 18   play--ut but at the outset one can try to predict

 

 19   and think about a definition of significant as

 

 20   being that which has the potential to help children

 

 21   with cancer.

 

 22             The second thing that really matters

 

 23   before a clinical trial begins is a risk/benefit

 

 24   assessment.  I think in the next several slides I

 

 25   will talk more about what counts as risk and what

 

                                                                32

 

  1   counts as benefit.

 

  2             Finally, it is a study design that will

 

  3   answer the question and that also does not

 

  4   subjugate the interests of any single subject to

 

  5   the overall needs of the research.  Again, embedded

 

  6   there are a couple of important ethical principles

 

  7   that I think are perhaps specific--at least the

 

  8   second one under study design--specific to research

 

  9   with a vulnerable population and, as Dr. Hirschfeld

 

 10   said, children certainly are considered and should

 

 11   be considered.

 

 12             The next slide, please.  This slide shows

 

 13   the criteria for the 405 category.  As I think

 

 14   everybody is aware, there are four categories of

 

 15   research that can be approved by IRBs under subpart

 

 16   D.  Almost all cancer research I think is approved

 

 17   under 405, that is, pediatric cancer research.  It

 

 18   is research that involves more than minimal risk

 

 19   but presents the prospect of direct benefit to the

 

 20   individual subject if the risk is justified by the

 

 21   anticipated benefit to the subject; if the

 

 22   risk/benefit ratio is less than or equal to the

 

 23   alternatives; and if parental permission and assent

 

 24   are obtained.

 

 25             The next slide, please.  As we weigh risk

 

                                                                33

 

  1   and benefit in research ethics, it is important to

 

  2   remember that risk means risk to the subject but

 

  3   benefit may include benefits to the subject,

 

  4   benefits to other patients, benefits to society or

 

  5   benefits to an investigator or a sponsor.  I think

 

  6   what we are aiming for in research involving

 

  7   children in some sense is limiting the benefits

 

  8   that we think about in a risk/benefit analysis so

 

  9   that the benefits that come to the subject are the

 

 10   ones that we are thinking about as we weigh risk to

 

 11   the subject, and that we avoid a situation where

 

 12   children are used as a means to an end.  To go back

 

 13   to Emmanuel Kant and the idea that children are

 

 14   valued and protected, I think it is inherent in

 

 15   this sort of balancing.

 

 16             The next slide, please.  This is a slide

 

 17   that looks at some of the issues in early drug

 

 18   development involving children with cancer.  There

 

 19   has been a controversy over, what I have put in

 

 20   quotes here, therapeutic intent.  The point here is

 

 21   that the prospect of direct benefit is the key

 

 22   ethical and regulatory issue and, in my view, a

 

 23   percentage view of what that potential for

 

 24   therapeutic intent might be isn't that important.

 

 25   That is, I think even a very low chance of

 

                                                                34

 

  1   therapeutic benefit for the child should count as a

 

  2   prospect of direct benefit to the child.  Again, my

 

  3   interpretation of the word prospect is a very broad

 

  4   one, admittedly, but this is where the issue of

 

  5   interpretation comes in.  As the discussion goes

 

  6   on, we can talk about how prospect ought to be

 

  7   interpreted.

 

  8             The second bullet point you see on this

 

  9   slide has, in parentheses, the potential for 405

 

 10   creep, that is, moving this issue of commensurate

 

 11   experience that children with cancer have already

 

 12   been through a lot so that it is okay to put them

 

 13   through one more thing.  This doesn't stand up in

 

 14   my view as a valid justification for exposing

 

 15   children with cancer to risk.

 

 16             The alternatives is another key issue that

 

 17   is discussed, if you recall, in the 405 criteria.

 

 18   There needs to be favorable outcome for the child

 

 19   compared to the alternatives.

 

 20             The next slide, please.  If we are on the

 

 21   same page, this should be a slide that says options

 

 22   on top.  It has at least three different pathways

 

 23   that families and children can seek out when a

 

 24   child has refractory, untreatable cancer.  On your

 

 25   left is a Phase I study; in the middle is

 

                                                                35

 

  1   alternative medicine and on the right is hospice

 

  2   philosophy care.

 

  3             The next slide shows further

 

  4   considerations regarding Phase I oncology research

 

  5   in children.  The first is to point out that

 

  6   subject selection is not a major controversy in

 

  7   this realm, that is to say, Phase I studies are

 

  8   done involving healthy children but it is not an

 

  9   issue of wanting to do Phase I cancer research on

 

 10   healthy children.  That, to my knowledge, is not a

 

 11   controversy but I put it here because it is

 

 12   important to try to contextualize pediatric cancer

 

 13   research in the broader picture of research

 

 14   involving children.  As I said before, I think that

 

 15   Phase I research qualifies, in my mind, as research

 

 16   with the prospect or direct benefit.

 

 17             Most importantly on this slide, is that

 

 18   potential for benefit mitigates but does not

 

 19   eliminate the need for protection from research

 

 20   risk.  To be more clear about that, it is the

 

 21   potential for benefit that is balanced against the

 

 22   risk that mitigates it, but I think the charge to

 

 23   the committee and the work we are going to do this

 

 24   morning is still extremely important.  The need for

 

 25   protection from research risk is not eliminated by

 

                                                                36

 

  1   the potential for benefit.

 

  2             The next slide, please.  This points out

 

  3   some issues around alternative medicine.  The

 

  4   reason that I put this here is that I think there

 

  5   is a yardstick of fairness that we need to keep in

 

  6   mind.  It is often the case that when research is

 

  7   being done it is held to a higher standard or a

 

  8   different standard than what is happening in the

 

  9   non-research world, and it is very important I

 

 10   think to the families and the children involved

 

 11   that we try to put this in the lens that they are

 

 12   viewing this off from, and to make it difficult to

 

 13   access research or to have children participate in

 

 14   well-designed, safely monitored research, in some

 

 15   ways, runs the risk of shunting them to alternative

 

 16   medicine where there are vulnerability concerns.

 

 17   It is very prevalent phenomena for children with

 

 18   refractory cancer.  I think there are major ethical

 

 19   differences when it comes to children getting

 

 20   alternative therapy compared to adults who can make

 

 21   their own decision.  I think we have a very

 

 22   important obligation to prevent harm when it comes

 

 23   to children who are getting alternative medicine,

 

 24   and I think it is extremely important that

 

 25   alternative medicine possibilities be studied in a

 

                                                                37

 

  1   rigorous and careful way.  But the bottom line is

 

  2   that we need to communicate with families and

 

  3   children.  The ones that the research community

 

  4   encounters may also be taking alternative medicine

 

  5   and if we don't know what medications are being

 

  6   taken, then we won't have the ability to study drug

 

  7   interaction with alternative medications and the

 

  8   experimental agent, for example.  I just think that

 

  9   it is very important that we keep alternative

 

 10   medicine in mind as something that is out there and

 

 11   we shouldn't be blind to it.

 

 12             The next slide, please.  This slide has a

 

 13   few words about hospice care for children who have

 

 14   refractory disease.  Now, some people I think have

 

 15   the experience that those who come to Phase I

 

 16   studies are self-referred, not interested in

 

 17   hospice philosophy care, wanting to continue to

 

 18   pursue anti-neoplastic therapy but, in my

 

 19   experience, that is not the case.  In fact, many

 

 20   families who seek Phase I studies also are amenable

 

 21   to having their child get hospice philosophy care.

 

 22   So, the two are not incompatible.  I think it is an

 

 23   under-developed approach in children.  It is not

 

 24   the main focus of what we are here about today but

 

 25   I felt that it would be incomplete to give this

 

                                                                38

 

  1   talk without mentioning that hospice philosophy

 

  2   care should be part of the consent process for

 

  3   Phase I studies.

 

  4             The next slide, please.  This moves from

 

  5   what really matters before the conduct of the trial

 

  6   to during the conduct of the trial.  The three

 

  7   items that really matter during the conduct of the

 

  8   trial are informed consent which, in my view, is a

 

  9   communication process in addition to the

 

 10   documentation that happens; ongoing monitoring via

 

 11   a data safety monitoring board, if appropriate, and

 

 12   I understand that much of the discussion later on

 

 13   will have to do with when it is appropriate and

 

 14   when it is not necessary; and ethical action to

 

 15   suspend or stop a study at the right time.  It is

 

 16   easier said than done but in parentheses I thought

 

 17   I would say not too soon but not too late either.

 

 18   So, the question of when a study should be

 

 19   suspended or stopped is a key ethical question that

 

 20   happens during the conduct of a study and whether a

 

 21   study needs to be stopped at all.  I guess in most

 

 22   cases there is no need to stop it but that question

 

 23   needs to be always asked in the same way house

 

 24   officers always need to ask themselves does this

 

 25   child need a spinal tap.  It is a question that is

 

                                                                39

 

  1   part of the monitoring process as an embedded

 

  2   function.

 

  3             The next slide shows the Nuremberg code.

 

  4   This is a quick bit about informed consent.  The

 

  5   Nuremberg code said that the voluntary consent of

 

  6   the human subject is absolutely essential.  These

 

  7   are slides that I have shown at previous meetings

 

  8   so I think we can go fairly quickly through them.

 

  9             The next slide asks the rhetorical

 

 10   question of whether we can do any pediatric

 

 11   research at all, and just points out that if the

 

 12   answer is no, that is, if we have to adhere to

 

 13   strict interpretation of the Nuremberg or literal

 

 14   rather than in the spirit of the law

 

 15   interpretation, children as a group will suffer.

 

 16   You saw in the Belmont quotation earlier that there

 

 17   is a clear recognition that there needs to be some

 

 18   research involving children so that we can both

 

 19   protect children adequately but be sure that we

 

 20   make progress in childhood disease.

 

 21             The next slide talks about three ways of

 

 22   respecting Nuremberg and still doing pediatric

 

 23   research by using parents as surrogates and

 

 24   obtaining parental permission; by involving

 

 25   children when appropriate and obtaining their

 

                                                                40

 

  1   assent; and by providing societal protection with

 

  2   IRB approval as the most obvious but also meetings,

 

  3   similar to what we are doing this morning,

 

  4   investigator integrity and other things that

 

  5   provide societal protection for children, we can, I

 

  6   think, ethically do pediatric research.

 

  7             The next slide shows the difference

 

  8   between parental permission and informed consent

 

  9   and, again, says that the autonomous authorization

 

 10   of an adult--the difference between adult and

 

 11   pediatric ethics is more robust than a proxy

 

 12   decision and points out, from the Academy of

 

 13   Pediatrics, that the responsibilities of a

 

 14   pediatrician to his or her patient exist

 

 15   independent of parental desires or proxy consent.

 

 16   I think that there is a congruent statement that

 

 17   one could make here that says that an

 

 18   investigator's responsibility to his or her subject

 

 19   exists independent of parental desires or proxy

 

 20   consent.

 

 21             The next slide shows that parental

 

 22   permission is not the oral equivalent of informed

 

 23   consent, and that surrogate decision-making is

 

 24   necessarily less authentic.  I am going to skip

 

 25   past the next slide which shows proxy consent,

 

                                                                41

 

  1   substituted judgment and best interests, because I

 

  2   think this is familiar ground for most people and

 

  3   we have already emphasized best interests.

 

  4             I will go to a slide that says informed

 

  5   consent in pediatrics equals parental permission

 

  6   and the assent of the child.  Here I want to say

 

  7   that the combination of those two can potentially

 

  8   be more powerful, if done right, than an

 

  9   individual.  This has to do with family centered

 

 10   ethics that really seek to care for and do

 

 11   effective communication with a family, which is a

 

 12   dynamic and challenging process, admittedly.  But I

 

 13   think both of these issues are very important.

 

 14             The next slide, please.  This provides the

 

 15   regulatory definition of assent, which is a child's

 

 16   affirmative agreement to participate in research.

 

 17   The key point here is that mere failure to object

 

 18   should not be construed as assent.  That is, the

 

 19   silence of an older child for research

 

 20   participation can't be interpreted as their assent.

 

 21   Again, there is room for regulatory interpretation

 

 22   here.  There is a great deal of controversy around

 

 23   assent and requirements for assent, and I think

 

 24   there is likely to be a fair amount of variability

 

 25   across IRBs with regard to this issue and I would

 

                                                                42

 

  1   be happy to discuss this further during our

 

  2   discussion.

 

  3             The next slide, please.  This slide shows

 

  4   some differences between assent in the clinical and

 

  5   research context, and points out the fact that

 

  6   research is supererogatory, that is, as opposed to

 

  7   a clinical context where there is a strong best

 

  8   interests argument to be made.  Generally speaking,

 

  9   in research the decision is more voluntary and, for

 

 10   that reason, assent is more powerful phenomenon, in

 

 11   my view, ethically speaking in research than it

 

 12   would be in the clinical context.

 

 13             The bottom bullet point here is also

 

 14   important I think as a principle perhaps for us to

 

 15   consider, and that is the older the child, the more

 

 16   assent contributes to the ethical justification for

 

 17   the study.  This is a problem for diseases that

 

 18   happen in younger children certainly but, all

 

 19   things being equal, an older child I think who can

 

 20   participate in the decision gives us more ethical

 

 21   justification for proceeding in research endeavors.

 

 22             The next slide just points out a piece of

 

 23   data.  This is a scale that we did in our study of

 

 24   informed consent about decision-making preference.

 

 25   It shows everything from, number one, a parent who

 

                                                                43

 

  1   wants to leave all decisions to the doctor and

 

  2   perhaps to an investigator, and then a continuum to

 

  3   number five, a parent who wants to make final

 

  4   selection about which treatment their child will

 

  5   receive.

 

  6             The next slide shows a sample of 108

 

  7   parents.  The reason that I included it this

 

  8   morning is to point out the variability among

 

  9   parents and families when it comes to how they want

 

 10   to make decisions.  You see in this slide a large

 

 11   number of parents in the middle, within the green,

 

 12   red and grey columns, who fit into a shared

 

 13   decision-making model.  In my view, this is why

 

 14   informed consent is important during the conduct of

 

 15   research.  Most people want a shared

 

 16   decision-making approach whether it comes to

 

 17   treatment or research participation and

 

 18   communication.  Effective communication is really

 

 19   the key issue for informed consent.

 

 20             The next slide.  As I wind down the talk

 

 21   and get to the conclusion, I want to make the point

 

 22   that the over-interpretation of regulatory concerns

 

 23   can prevent the ethically meaningful participation

 

 24   of children in research.

 

 25             Can you still hear me?

 

                                                                44

 

  1             MS. CLIFFORD:  We can still hear you.

 

  2             DR. KODISH:  Great!  I heard a beep on the

 

  3   phone.  I am going to tell a quick story to

 

  4   illustrate this point.  Heather K was diagnosed

 

  5   with a vaginal rhabdomyosarcoma at a children's

 

  6   hospital in the Midwest within the past few months.

 

  7   At diagnosis, Heather had a tumor that was causing

 

  8   intestinal compression.  Her pediatric oncologist

 

  9   talked to the family about the diagnosis and then

 

 10   subsequently discussed a Phase III non-randomized

 

 11   study sponsored by the IRS/COG.  The family

 

 12   provided informed consent and signed a document at

 

 13   6:05 p.m.  The plan was to begin chemotherapy the

 

 14   following day but the patient developed a bowel

 

 15   obstruction at 11:00 p.m. and chemotherapy was

 

 16   emergently started.  At midnight nothing happened

 

 17   that was ethically significant.  Clinically, the

 

 18   patient was continuing to get her chemotherapy.

 

 19   But the next morning, when the CRA, the data

 

 20   person, came to enroll Heather in this Phase III

 

 21   study, the RDE, or the remote data entry system,

 

 22   made enrollment impossible.  The reason that

 

 23   enrollment was impossible was that the date

 

 24   chemotherapy was started was the previous date and

 

 25   the form would not permit enrollment to happen if

 

                                                                45

 

  1   chemotherapy had already been started.

 

  2             So, what was a well-intentioned regulation

 

  3   system designed to prevent people from being

 

  4   entered on study if consent had not yet been

 

  5   obtained--in fact, in this case everything went

 

  6   perfectly from an ethical perspective but the

 

  7   patient was not allowed to be entered on study.  I

 

  8   think that this is a cautionary tale and I wanted

 

  9   to bring it to the attention of the panel today.

 

 10             Next slide, please.  We see many

 

 11   well-intentioned regulatory protections and it is

 

 12   important to realize that they can paradoxically

 

 13   prevent the ethical participation of children in

 

 14   cancer research and Heather's story is one example

 

 15   of that.  The physician then needed to go back to

 

 16   the family and explain that, unfortunately, we

 

 17   weren't able to include her as a subject in the

 

 18   research.  It wasn't going to change her treatment

 

 19   at all but the future treatment of children with

 

 20   rhabdomyosarcoma in some ways is harmed by the fact

 

 21   that this regulatory mechanism prevented Heather

 

 22   from being a subject in the study.  The only

 

 23   alternative would have been for the person doing

 

 24   remote data entry to fabricate and to say that the

 

 25   date chemotherapy was started was the day that she

 

                                                                46

 

  1   was being entered on study, and that would have,

 

  2   number one, been an unethical lie and, number two,

 

  3   would have been picked up on an audit if the

 

  4   subject had been audited subsequently though it may

 

  5   have been, in fact, the ethical thing to do because

 

  6   consent was obtained in an appropriate way, it is

 

  7   an important study, and all of the things that we

 

  8   have bee talking about, but the regulatory

 

  9   apparatus prevented an ethical action from taking

 

 10   place and I think it is a disturbing story.

 

 11             The next slide shows a synergistic

 

 12   approach.  The protection of human subjects has

 

 13   been done both through education and regulation and

 

 14   we need to be concerned about developing too much

 

 15   regulation at the expense of education and the

 

 16   expense of thoughtful ethical action.

 

 17             The next slide just has a few quick points

 

 18   about what matters after a trial is closed.

 

 19   Monitoring for late effects of therapy is an

 

 20   important ethical issue after a trial has closed.

 

 21   The publication of results and dissemination of

 

 22   findings is ethically important.  If the science

 

 23   isn't disseminated, then it is like a tree falling

 

 24   in a forest that nobody hears.  Finally, the return

 

 25   of results to the subjects who participated is an

 

                                                                47

 

  1   ethically under-looked and I think very important

 

  2   issue that symbolizes the partnership that we have

 

  3   with subjects and their families, and I think we

 

  4   need to do a better job than we are doing currently

 

  5   after a trial has closed in getting results back to

 

  6   the subjects.

 

  7             The next slide shows conceptually the main

 

  8   balance as a point of conclusion in pediatric

 

  9   research ethics, that the best interests of the

 

 10   child-subject are, in fact, balanced against

 

 11   science to benefit others and we need to be

 

 12   cognizant of that balance at all times and be sure

 

 13   that the best interests of the child are not

 

 14   subjugated.

 

 15             The next slide shows a couple of

 

 16   conclusions.  The first is that beneficence, as

 

 17   described in the Belmont report, is the key ethical

 

 18   principle that I believe should guide monitoring of

 

 19   patients in studies.  Also, a risk/benefit

 

 20   assessment by the investigator, by the IRB and by

 

 21   others perhaps is more important than informed

 

 22   consent, and that is because I don't think informed

 

 23   consent has the ethical importance in pediatrics

 

 24   that it does in adult medicine, and also because of

 

 25   the relatively ineffective communication process

 

                                                                48

 

  1   that is currently happening with informed consent.

 

  2   I would be happy to talk more about that in the

 

  3   discussion.

 

  4             The next slide shows that the protection

 

  5   of children from research risk and the imperative

 

  6   to improve childhood cancer treatment are both

 

  7   ethically important.  The bottom point here is that

 

  8   regulatory fervor intended to protect children

 

  9   currently threatens the ethical conduct of

 

 10   pediatric cancer research, as I tried to illustrate

 

 11   in Heather's story, and we need to remember, I

 

 12   think, that there is an ethical imperative to do

 

 13   work in childhood cancer to improve the care of

 

 14   children with cancer.

 

 15             The final slide points out that children

 

 16   are both vulnerable subjects who need protection

 

 17   from research risk and a neglected class--and they

 

 18   continue to be a neglected class despite our best

 

 19   efforts--that need better access to the benefits of

 

 20   research.

 

 21             I thank you all for tolerating the virtual

 

 22   reality nature of this talk and hope that I have

 

 23   been able to make a contribution.  Thank you.

 

 24             DR. SANTANA:  Thanks, Eric.  Eric, are you

 

 25   planning to stay on line for the rest of the

 

                                                                49

 

  1   morning?

 

  2             DR. KODISH:  I am.  The only question is

 

  3   whether I should do it by phone or by Webcast.

 

  4             DR. SANTANA:  Okay, because if you are

 

  5   going to stay, then we will just hold the questions

 

  6   for the general discussion, if that is okay with

 

  7   you.

 

  8             DR. KODISH:  That is fine.

 

  9             DR. SANTANA:  But I do want you to stay on

 

 10   the phone line, if at all possible, for the

 

 11   discussion because I think we can communicate

 

 12   better that way.

 

 13             DR. KODISH:  Okay, what I will try to do

 

 14   is watch but mute the sound.

 

 15             DR. SANTANA:  That is fine.

 

 16             DR. KODISH:  Thank you, Victor.

 

 17             DR. SANTANA:  Okay, good.  I also want to

 

 18   thank John for advancing your slides on your

 

 19   behalf.  Dr. Carome, you are next.

 

 20         Legal Responsibilities for HHS Supported Studies

 

 21             DR. CAROME:  Good morning.  I would like

 

 22   to thank the subcommittee members for inviting me

 

 23   to give a brief presentation on legal

 

 24   responsibilities for studies conducted and

 

 25   supported I think originally by the federal

 

                                                                50

 

  1   government and since I speak on behalf of HHS, I

 

  2   have limited it to HHS, the Department of Health

 

  3   and Human Services.

 

  4             What I am quickly going to do is go over,

 

  5   first of all, the applicability of our regulations.

 

  6   Then I am going to talk very quickly about the

 

  7   major requirements of 45 CFR Part 46, Subpart A,

 

  8   which are the general protections for human subject

 

  9   research.  Then I am going to finish up by talking

 

 10   about the major requirements of 45 CFR, part 46,

 

 11   Subpart D, which are the additional protections for

 

 12   children involved as subjects in research.

 

 13             Again, the regulations I am referencing,

 

 14   45 CFR Part 46, are the HHS regulations for the

 

 15   protection of human subjects.  They have four

 

 16   subparts.  The regulations were last revised in

 

 17   2001.  One of the subparts, Subpart B, was revised

 

 18   at that point but most of the regulations remain

 

 19   the same as when they were promulgated more than

 

 20   two decades ago.

 

 21             So, what is the applicability of these

 

 22   regulations?  Our regulations apply in two

 

 23   circumstances.  The most common is research

 

 24   conducted or supported by the Department that are

 

 25   not otherwise exempt.  That includes clinical

 

                                                                51

 

  1   trials conducted intramurally by the NIH or funded

 

  2   by the NIH, as well as many other agencies within

 

  3   the Ddpartment.  A second way in which research can

 

  4   be covered by these regulations is research that is

 

  5   conducted at an institution holding an applicable

 

  6   assurance of compliance approved by our office.

 

  7   So, any institution that receives funding from our

 

  8   Department to conduct human subject research must

 

  9   execute a written agreement in which the

 

 10   institution pledges to comply with our regulations,

 

 11   and in that document many institutions voluntarily

 

 12   extend the same regulations to all research

 

 13   regardless of sponsorship.  In doing so, the

 

 14   assurance comes to cover privately sponsored

 

 15   research.

 

 16             This slide demonstrates the relationship

 

 17   and the overlap between the applicability of our

 

 18   regulations and the FDA regulations.  You can see

 

 19   that there is in the middle an overlap.  The

 

 20   overlap may occur in two circumstances.  One is

 

 21   where NIH sponsors a clinical trial or other

 

 22   clinical research, or any research, that involves

 

 23   an FDA-regulated test article.  Another

 

 24   circumstance is where an institution, holding an

 

 25   assurance with our office in which they voluntarily

 

                                                                52

 

  1   agreed to extend that assurance to all research, is

 

  2   engaged in an industry, privately sponsored

 

  3   research, project involving an FDA-regulated test

 

  4   article.

 

  5             Very quickly, what are the major

 

  6   provisions of Subpart A?  As was previously noted,

 

  7   the regulations, we believe, are clearly founded

 

  8   upon an ethical framework that was articulated in

 

  9   the Belmont report.  Its three basic ethical

 

 10   principles, and the fundamental provisions of the

 

 11   regulations can be divided in three groups.  One is

 

 12   the provisions related to and assurance of

 

 13   compliance.  The second is those related to the IRB

 

 14   requirements, institutional review boards, and the

 

 15   third is those requirements related to legally

 

 16   effective informed consent.

 

 17             With respect to assurances, the

 

 18   regulations stipulate that each institution engaged

 

 19   in research covered by the regulations and which is

 

 20   conducted or supported by the Department shall

 

 21   provide assurance satisfactory to the HHS Secretary

 

 22   that it will comply with the requirements set forth

 

 23   in the regulations.

 

 24             The regulations further stipulate specific

 

 25   elements that must be part of an assurance.  There

 

                                                                53

 

  1   must be a statement of principles governing the

 

  2   institution in the discharge of its

 

  3   responsibilities for protecting the rights and

 

  4   welfare of human subjects.  And, the regulations

 

  5   state that those principles must apply to all

 

  6   research regardless of whether or not it is covered

 

  7   by the assurance.

 

  8             The assurance must designate at least one,

 

  9   and many institutions designate more than one,

 

 10   institutional review board and that must include a

 

 11   list of the IRB members and their relative

 

 12   capacities, and there must be a reference to

 

 13   written IRB procedures.  There are requirements

 

 14   related to the IRB and they include specification

 

 15   of what the IRB membership must include, such as at

 

 16   least one person whose primary interests are in the

 

 17   scientific area and at least one member whose

 

 18   primary interests are in a non-scientific area, and

 

 19   at least one member who is not otherwise affiliated

 

 20   with the institution or a member of a family

 

 21   affiliated with the institution.

 

 22             The regulations have specific provisions

 

 23   related to how the IRB should function and operate;

 

 24   when it must conduct review in terms of initial and

 

 25   continuing review.  Then there are provisions

 

                                                                54

 

  1   related to expedited review for certain categories

 

  2   of minimal risk research and there are detailed

 

  3   lists of specific criteria an IRB must find in

 

  4   order to approve research.  For example, the

 

  5   regulations state that in order to approve research

 

  6   an IRB must find that the risks to the subjects are

 

  7   minimized and reasonable in relationship to the

 

  8   anticipated benefits, if any, to the subjects and

 

  9   the knowledge that is to be gained.  Then, there

 

 10   are other provisions for the records that an IRB

 

 11   must maintain.

 

 12             The last set or provisions in Subpart A

 

 13   deal with legally effective informed consent.  They

 

 14   include an introductory paragraph that talks about

 

 15   the general requirements.  For instance, no

 

 16   investigator may involve a human subject in

 

 17   research unless the informed consent of the subject

 

 18   or a legally authorized representative of the

 

 19   subject has been obtained, except in certain

 

 20   limited circumstances in which informed consent can

 

 21   be waived.

 

 22             The regulations go on to stipulate basic

 

 23   elements that I think most people are familiar

 

 24   with: the nature of the research; the reasonably

 

 25   foreseeable risks; the reasonably foreseeable

 

                                                                55

 

  1   benefits, if any, to the subject; and others, such

 

  2   as alternatives that a subject may choose instead

 

  3   of entering the research.  The regulations

 

  4   stipulate that consent must generally be

 

  5   documented, except in some limited circumstances.

 

  6   Then, there are waiver provisions both for

 

  7   obtaining informed consent at all or for documented

 

  8   informed consent, and I won't go into those in

 

  9   detail.

 

 10             Let's turn finally to the provisions for

 

 11   research involving children under Subpart D, the

 

 12   additional protections for children.  Again, this

 

 13   is a subpart that is unique to the Department of

 

 14   Health and Human Services.  Whereas all the Subpart

 

 15   A provisions that I just went over have been

 

 16   adopted by other departments and agencies, Subpart

 

 17   D has only been adopted by the Department of

 

 18   Education in addition to our department.

 

 19             Subpart D applies to all research

 

 20   involving children as subjects conducted or

 

 21   supported by our department.  It is important to

 

 22   note that there is a specific definition of

 

 23   children in the regulations, and they are persons

 

 24   who have not attained the legal age for consent to

 

 25   treatments or procedures involved in the research

 

                                                                56

 

  1   under the applicable law of the jurisdiction in

 

  2   which the research will be conducted.  It is

 

  3   important to note that in order to then understand

 

  4   who a child is with respect to the research

 

  5   regulations, you must understand state and local

 

  6   law that defines who can consent to what and at

 

  7   what age.  Therefore, a child in one state might

 

  8   not be a child in another state for the purposes of

 

  9   these regulations.

 

 10             The Subpart D requirements in

 

 11   general--first of all, you have to satisfy all the

 

 12   requirements of Subpart A.  So, if a research

 

 13   project involving children doesn't satisfy some

 

 14   provision of Subpart A, then it is moot about the

 

 15   additional provisions.  The research would not be

 

 16   approvable.  But if the research is approvable

 

 17   under Subpart A, there are additional requirements

 

 18   of Subpart D which must be fulfilled and satisfied.

 

 19             As Eric referenced, there are four

 

 20   categories of research that are approvable under

 

 21   Subpart D under our regulations.  These are

 

 22   primarily scaled to risk versus benefit as you walk

 

 23   through each of these categories, and I am going to

 

 24   do that very quickly.

 

 25             The first category, 404, is research not

 

                                                                57

 

  1   involving greater than minimal risk, and minimal

 

  2   risk is defined in Subpart A.  In order for this

 

  3   research to be approved under this category, an IRB

 

  4   must make one general finding.  It must find that

 

  5   there are adequate provisions for soliciting the

 

  6   assent of the child and permission of the parents

 

  7   or guardians, as set forth in Section 408.

 

  8             The next category, Section 405, which Eric

 

  9   went into more detail, is research involving

 

 10   greater than minimal risk but presenting the

 

 11   prospect of direct benefit to the individual

 

 12   subjects.  So, the benefit has to be tied to the

 

 13   subjects as opposed to society in general and the

 

 14   knowledge to be gained.  Here, the IRB must make

 

 15   three specific findings.  The IRB must find that

 

 16   the risk is justified by the anticipated benefits

 

 17   to the subject; the relationship of the anticipated

 

 18   benefit to the risk is at least as favorable to the

 

 19   subjects as that presented by available

 

 20   alternatives outside the research context; and,

 

 21   again, the same provisions for assent and

 

 22   permission apply throughout these four categories.

 

 23             The next category, 406, involves greater

 

 24   than minimal risk and no prospect of direct benefit

 

 25   to the individual subjects, but likely to yield

 

                                                                58

 

  1   generalizable knowledge about the subject's

 

  2   disorder or condition.  For this category there are

 

  3   four criteria that an IRB must find.  They must

 

  4   find that, first, that the risk represents a minor

 

  5   increase over minimal risk.  Whereas minimal risk

 

  6   is defined in the regulations, what a minor

 

  7   increase means is not defined so that is left up to

 

  8   the judgment of the IRBs.

 

  9             Next, the IRB must find that the

 

 10   intervention or procedure within the research

 

 11   presents experiences to the subjects that are

 

 12   reasonably commensurate with those inherent in the

 

 13   actual or expected medical, dental, psychological,

 

 14   social or educational situation of the child.

 

 15   Commensurability is one of the factors that Eric

 

 16   touched on but applies only in this category, 406.

 

 17             The next two provisions--the IRB must find

 

 18   under 406 that the intervention or procedure is

 

 19   likely to yield generalizable knowledge about the

 

 20   subject's disorder or condition which is of vital

 

 21   importance for the understanding or amelioration of

 

 22   the subject's disorder or condition.  I think the

 

 23   key words here are that you have to understand that

 

 24   the child must have a disorder or condition, two

 

 25   terms that are not otherwise defined in the

 

                                                                59

 

  1   regulation and are of vital importance.  So, it is

 

  2   sort of a higher standard than the usual

 

  3   generalizable knowledge standard that probably

 

  4   applies to research under Subpart A only.  Lastly

 

  5   is the assent or permission provisions.

 

  6             The fourth category and final category is

 

  7   research that is not otherwise approvable under one

 

  8   of these four categories which presents a

 

  9   reasonable opportunity to understand, prevent or

 

 10   alleviate a serious problem affecting the health or

 

 11   welfare of children.  For this, the IRB still must

 

 12   review and assess the research with respect to

 

 13   Subpart A and D, and must find that the research

 

 14   presents a reasonable opportunity to further the

 

 15   understanding, prevention or alleviation of a

 

 16   serious health problem affecting the health or

 

 17   welfare of children.

 

 18             The project is then forwarded to the

 

 19   Department.  They come through our office and we

 

 20   act on behalf of the Secretary to process these.

 

 21   In order for the research then to be approved, the

 

 22   Secretary, after consultation with a panel of

 

 23   experts in pertinent disciplines and following an

 

 24   opportunity for public review and comment, must

 

 25   determine either that the research in fact

 

                                                                60

 

  1   satisfies one of the other three categories, 404,

 

  2   405 or 406 or, if not, three things must be met:

 

  3   that research presents a reasonable opportunity

 

  4   standard that I previously went over; that the

 

  5   research will be conducted in accordance with sound

 

  6   ethical principles, and hopefully that is something

 

  7   that applies to all research conducted; and

 

  8   adequate provisions for the assent of the child and

 

  9   parental permission.

 

 10             Finally, there are some additional

 

 11   provisions of Subpart D that are provisions related

 

 12   to soliciting assent, and assent is not always

 

 13   required and an IRB may determine it is not

 

 14   warranted, particularly under category 405.  There

 

 15   are provisions for soliciting permission of

 

 16   parents, and the regulations speak to whether you

 

 17   need both parents' permission.  If the category is

 

 18   405 one parent's permission is sufficient but for

 

 19   406 or 407 two parents are required, except in very

 

 20   limited circumstances.

 

 21             It is important to note that there are

 

 22   provisions for waiving parental permission or

 

 23   guardian permission.  Just like informed consent

 

 24   can be waived under Subpart A for research

 

 25   involving adults, parental permission can be waived

 

                                                                61

 

  1   in certain circumstances and this is I think unique

 

  2   to our regulations and not found in the parallel

 

  3   regulations within the FDA.

 

  4             Finally, there are specific protections

 

  5   for subjects who are wards of the state or any

 

  6   other agency, institution or entity for research

 

  7   approved under 406 or 407.  Among those

 

  8   requirements, there must be a specific advocate

 

  9   appointed for each child who is participating in

 

 10   such research who is a ward.

 

 11             In summary, I have quickly tried to go

 

 12   over the applicability of our regulations and

 

 13   contrasted that with the FDA regulations

 

 14   applicability.  I have gone over the major

 

 15   requirements of Subpart A of our regulations and

 

 16   finished up with a discussion of Subpart D, and I

 

 17   thank you for your attention.

 

 18             DR. SANTANA:  Thanks, Dr. Carome.  Dr.

 

 19   Hirschfeld?

 

 20             Legal Responsibilities for Studies with

 

 21                      FDA Regulated Products

 

 22             DR. HIRSCHFELD:  I would also like to

 

 23   thank Dr. Carome and note that when he was wearing

 

 24   a uniform which was a color more consistent with

 

 25   the theme of the day, he was the head of the IRB at

 

                                                                62

 

  1   Walter Reed Army Medical Center.  I also want to

 

  2   thank him for his efforts on clarification of the

 

  3   regulations in ongoing discussions as they apply to

 

  4   pediatric oncology, and he has taken a leadership

 

  5   role in the Office for Human Research Protection in

 

  6   that regard.

 

  7             I am going to even more quickly, I hope,

 

  8   go through the FDA regulations.  One might ask what

 

  9   is a pediatric oncologist doing talking about FDA

 

 10   regulations, but that is one of the strengths of

 

 11   the FDA, that there are wonderful opportunities to

 

 12   be involved in many aspects or research in clinical

 

 13   medicine, including the development of regulations.

 

 14   I was on the working group that developed the

 

 15   Subpart D and, in fact, wrote the first draft of

 

 16   that document.

 

 17             As Dr. Carome pointed out, there is some

 

 18   overlap, and these slides have a lot of data which

 

 19   is intended for reference and I will not go through

 

 20   all the aspects of all the slides, but just to note

 

 21   that there are laws synonymous with an act or

 

 22   statute which are developed and passed by the

 

 23   Legislative Branch and signed by the President and

 

 24   these are published in the United States Code.

 

 25   Then there are regulations synonymous with rule,

 

                                                                63

 

  1   and these are developed and published by the

 

  2   Executive Branch, the various departments and

 

  3   agencies within the Executive Branch doing the

 

  4   detailed work, and these are published in the Code

 

  5   of Federal Regulations, which is referred to as the

 

  6   CFR.

 

  7             The FDA authority is derived from multiple

 

  8   laws and regulations, and the focus is on product

 

  9   and product use.  There are a number of applicable

 

 10   regulations for good clinical practice in the

 

 11   research setting, and these include the human

 

 12   subject protection, which is in 21 CFR, Part 50;

 

 13   financial disclosures, which is in Part 54;

 

 14   institutional review boards, which is in Part 56;

 

 15   and investigational new drugs, which is in part

 

 16   312.

 

 17             Part 50 has actually three sections to it.

 

 18   One is reserved for future use and Part D, you will

 

 19   notice, is the additional safeguards for children

 

 20   in clinical investigations, which is the focus of

 

 21   the discussion now.

 

 22   This is a catalog of all the various sections

 

 23   within Subpart D of 21 CFR, 50.  You will see that

 

 24   there is mapping and harmonization between the

 

 25   relevant sections of the HHS regulations.

 

                                                                64

 

  1             Now, the relationship--and this is just a

 

  2   textual representation of the schematic that Dr.

 

  3   Carome presented--is that FDA regulations apply to

 

  4   all research using FDA-regulated products.  In

 

  5   contrast, the HHS regulations apply to all research

 

  6   that is supported by HHS.  Research that is

 

  7   supported by HHS using FDA-regulated products is

 

  8   subject to both sets of regulations, and the

 

  9   regulations are harmonized although there are some

 

 10   differences which Dr. Carome elaborated on earlier.

 

 11   The definitions, you will see, parallel those

 

 12   definitions in the HHS regulations and put the onus

 

 13   of interpretation on the local jurisdiction and on

 

 14   the local IRBs, and that is the theme that persists

 

 15   throughout these regulations.  So, these

 

 16   definitions are included here to show that there is

 

 17   harmonization and in some cases, we believe, some

 

 18   clarification because the scope of FDA-regulated

 

 19   research is, in many ways, different and can apply

 

 20   to domains where HHS research is not applicable.

 

 21   So, it was important to have not only clarity on

 

 22   the definitions but consistency and, therefore,

 

 23   there are definitions that are included here so

 

 24   that there is not, we hope, much ambiguity in terms

 

 25   of how to apply and interpret these regulations at

 

                                                                65

 

  1   the local IRB level.

 

  2             Here, again, there is an emphasis on the

 

  3   concept that Eric Kodish developed for us a little

 

  4   earlier this morning, and that is children do not

 

  5   actually engage in a consent process.  Their

 

  6   parents provide permission for them to participate

 

  7   in the research.  Then, there is the same emphasis

 

  8   as in the HHS regulations that the child must at

 

  9   least be approached for assent.

 

 10             So, in addition to the other

 

 11   responsibilities assigned to IRBs, the FDA

 

 12   regulations ask that the IRB review clinical

 

 13   investigations involving children as subjects

 

 14   covered by Subpart D and approve only clinical

 

 15   investigations that satisfy the criteria which are

 

 16   described in Subpart 51, 52, 53 and the conditions

 

 17   of all other applicable sections of Subpart D.

 

 18             These are again mapped to the four risk

 

 19   categories which were developed in the 1970s and

 

 20   which, because of their serviceability and their

 

 21   flexibility, have been maintained to this date.

 

 22   These, again, discuss the concept of minimal risk

 

 23   here with specific examples of how it applies to

 

 24   pediatric research.

 

 25             Since the IRBs are a conduit through which

 

                                                                66

 

  1   research occurs, there are specific instructions on

 

  2   when IRBs may approve clinical investigations, and

 

  3   these are divided into the specific risk

 

  4   categories.  So, there is greater than minimal risk

 

  5   under 50.51.  In 50.52 there is greater than

 

  6   minimal risk presenting the prospect of direct

 

  7   benefit and the conditions, again, are analogous to

 

  8   the HHS regulations; and 50.53 shows that the IRBs

 

  9   can approve clinical investigations involving

 

 10   greater than minimal risk and no prospect of direct

 

 11   benefit but likely to yield generalizable knowledge

 

 12   about the subject's disorder or condition, and the

 

 13   same caveats about having a disorder or condition

 

 14   and having the prospect of generalizable knowledge

 

 15   apply, and these are addressed in some detail.

 

 16             In addition, there are IRB approval

 

 17   criteria which are explicitly stated and these

 

 18   include not only minimization of risk and that the

 

 19   risks are anticipated in relation to the benefit,

 

 20   but that the informed consent process is adequate

 

 21   and appropriately documented and looking for

 

 22   safeguards.  That is going to be theme which we are

 

 23   going to look at in detail, what safeguards can be

 

 24   and ought to be implemented.

 

 25             Subpart D addresses this explicitly. 

 

                                                                67

 

  1   There is a paragraph devoted to monitoring which I

 

  2   will quote briefly:  While the level of risk in a

 

  3   clinical investigation may change during the course

 

  4   of a study, appropriate strategies may be included

 

  5   in the study design that may mitigate risks.  These

 

  6   might include exit strategies in the case of

 

  7   adverse events or a lack of efficacy, or

 

  8   establishing a data monitoring committee to review

 

  9   ongoing data collection and recommend study

 

 10   changes, including stopping a trial on the basis of

 

 11   safety information.

 

 12             Part 56 addresses institutional review

 

 13   boards, and the general provisions and organization

 

 14   are discussed in the first part; IRB functions and

 

 15   operations in the second part; records and

 

 16   reporting in the fourth part; and the

 

 17   administrative actions for non-compliance in the

 

 18   fifth part.

 

 19             Now we come to the IND regulations, 312

 

 20   Subpart A, which are the general provisions which

 

 21   are outlined here.

 

 22             Subpart B, which are in essence the

 

 23   mechanics of an investigational new drug

 

 24   application and the obligations under those

 

 25   sections.

 

                                                                68

 

  1             Subpart C, which discusses the

 

  2   administrative actions, and Subpart D which goes

 

  3   into detail of the responsibilities of the sponsors

 

  4   and investigators.

 

  5             There is a Subpart E, which doesn't map

 

  6   explicitly to other HHS regulations, which

 

  7   addresses the drugs intended to treat

 

  8   life-threatening and severely debilitating

 

  9   illnesses which apply to pediatric oncology

 

 10   studies.  You will notice in the various paragraphs

 

 11   here that in 312.87 there is a requirement for

 

 12   active monitoring of conduct and evaluation of

 

 13   clinical trials.  It reads, for drugs covered under

 

 14   this section, the Commissioner and other agency

 

 15   officials will monitor the progress of the conduct

 

 16   and evaluation of clinical trials and be involved

 

 17   in facilitating their appropriate progress.  So,

 

 18   this places an FDA role in a dynamic way in the

 

 19   research being conducted in the realm of

 

 20   life-threatening illnesses.

 

 21             In addition, 312.88 has specific

 

 22   safeguards for patient safety which refer back to

 

 23   the other sections that were discussed, Parts 50,

 

 24   56, 312.  We didn't discuss 314 which is the NDA

 

 25   regulations and 600 which apply to the biologics

 

                                                                69

 

  1   but there are analogous regulations in these areas.

 

  2             I will just abstract from here that this

 

  3   includes the requirements for informed consent and

 

  4   institutional review boards, and that these

 

  5   safeguards further include the review of animal

 

  6   studies prior to initial human testing; the

 

  7   monitoring of adverse drug experience through the

 

  8   requirements of IND safety reports; safety update

 

  9   reports for marketing and postmarketing.

 

 10             So, our conclusions from this section are

 

 11   that the FDA has authority to regulate clinical

 

 12   studies using FDA-regulated products; that FDA

 

 13   regulations incorporate both IRB and FDA oversight

 

 14   of studies; that regulations exist for studies

 

 15   using products intended to treat life-threatening

 

 16   illnesses; and that regulations exist for providing

 

 17   additional safeguards for children enrolled in

 

 18   clinical investigations; and, as noted, HHS and FDA

 

 19   regulations are intended to be harmonized.  Thank

 

 20   you.

 

 21             DR. SANTANA:  Thank you, Dr. Hirschfeld.

 

 22   I think we will hold our questions until we

 

 23   reconvene at the point for discussion.  I think we

 

 24   are just a few minutes behind time.  We will take a

 

 25   15-minute break--Dr. Hirschfeld wants a 10-minute

 

                                                                70

 

  1   break.  We will take a 10-minute break and try to

 

  2   reconvene at almost 9:45.  Thank you.

 

  3             [Brief recess]

 

  4             DR. SANTANA:  We will go ahead and get

 

  5   started with the second part of the morning

 

  6   presentations.  To initiate that, Dr. Anderson,

 

  7   from CTEP, will be our next speaker.  Barry?  Eric,

 

  8   are you back on board?

 

  9             DR. KODISH:  I am here.

 

 10             DR. SANTANA:  Thank you, Eric.

 

 11             Enrollment and Monitoring Procedures for

 

 12                        NCI Funded Studies

 

 13             DR. ANDERSON:  I am Barry Anderson, from

 

 14   NCI CTEP, and I want to thank the FDA and Steven

 

 15   for inviting us to provide information about the

 

 16   enrollment and monitoring procedures for

 

 17   NCI-supported clinical trials.

 

 18             For pediatric cancer clinical trials, the

 

 19   appropriate enrollment of the individual patient,

 

 20   the child who is going to come onto the trial, as

 

 21   well as the monitoring of that individual patient's

 

 22   experience during the trial and the cumulative

 

 23   experience of all children who are involved in a

 

 24   clinical trial I think are necessary components in

 

 25   terms of trying to enhance the patient safety and

 

                                                                71

 

  1   the scientific validity of the trial itself.

 

  2             So, at the onset, from NCI's point of

 

  3   view, it is important to work to assure that each

 

  4   child accrued to a trial is receiving the

 

  5   appropriate treatment within the clinical trial

 

  6   itself, and that monitoring that is associated with

 

  7   the trial monitors the toxicity and effectiveness

 

  8   of the treatment intervention within each clinical

 

  9   trial both for that individual child, as well as

 

 10   for the trial overall.

 

 11             The words "safe" and "effective" can be

 

 12   applied to many of the standard treatments we use

 

 13   in pediatric oncology to treat various childhood

 

 14   cancers.  These words have special meaning in

 

 15   pediatric oncology.  As Dr. Kodish mentioned, there

 

 16   is a special sort of risk/benefit ratio that we

 

 17   always consider because, while therapy for

 

 18   childhood cancer is often successful and that is

 

 19   something that differs from much of medical

 

 20   oncology, the therapies that we use are always

 

 21   toxic in pediatric oncology and they always carry a

 

 22   risk of treatment-related morbidity and perhaps

 

 23   even death in many cases.

 

 24             So, selecting the proper treatment I think

 

 25   is essential because compared with other serious

 

                                                                72

 

  1   childhood diseases, such as asthma or cystic

 

  2   fibrosis, childhood cancer includes many distinct

 

  3   histologic diagnoses, and each tumor histology

 

  4   requires a distinct treatment appropriate with its

 

  5   own risks and benefits.  The chances of cure also

 

  6   diminish quickly if the proper therapy is not used

 

  7   at the outset.  That differs, I think, from some of

 

  8   the other more chronic diseases that are serious

 

  9   within childhood diseases but can have chances to

 

 10   change the therapeutic approach over time.

 

 11             In regards to enrollment, a question for

 

 12   the clinical trials done in pediatric oncology is

 

 13   who should be enrolled.  Pediatric oncology has

 

 14   evolved an approach of risk stratified treatment

 

 15   regimens and within each tumor histology the

 

 16   patient characteristics and the tumor

 

 17   characteristics establish a risk of relapse.  This

 

 18   risk of relapse then is used to stratify the

 

 19   treatment assignment for each child in terms of the

 

 20   type of clinical trial or the specific clinical

 

 21   trial they would be appropriate for.  Using this

 

 22   risk of relapse the intensity of the treatment that

 

 23   the child receives--and for intensity you can also

 

 24   say increased toxicity--is then set to best fit the

 

 25   child's cancer.  So, it is vital to treat the

 

                                                                73

 

  1   child, as best we can ascertain at the time they

 

  2   first present, according to the appropriate

 

  3   treatment regimen.

 

  4             By following this treatment stratification

 

  5   approach, the goal in pediatric oncology is to

 

  6   minimize the exposure to highly toxic therapies for

 

  7   those children who don't need that much treatment,

 

  8   in a relative sense, and also for the oncologists

 

  9   to have some comfort in knowing that another child

 

 10   who has a high-risk chance of relapse, that they

 

 11   will in fact potentially benefit from using a more

 

 12   intensive and more toxic treatment regimen.

 

 13             To apply this treatment stratification

 

 14   approach across an entire clinical trial, it is

 

 15   important that the eligibility criteria within the

 

 16   protocol by which all the patients are brought into

 

 17   the trial--that those protocol eligibility criteria

 

 18   are clear in regards to the clinical

 

 19   characteristics of the patient and the pathologic

 

 20   and biologic characteristics of the tumor--that all

 

 21   these characteristics are clear and easy to

 

 22   understand.

 

 23             The pediatric oncologists that are

 

 24   involved in the trial and who would be enrolling

 

 25   patients must be properly informed on how to apply

 

                                                                74

 

  1   the eligibility criteria that are presented in the

 

  2   eligibility section of the protocol itself.  If

 

  3   anyone has ever had experience in trying to bring a

 

  4   patient with rhabdomyosarcoma into a sarcoma trial,

 

  5   it can be a be very complicated endeavor and many

 

  6   mechanisms have been put in place to assist the

 

  7   pediatric oncologist to make sure that the proper

 

  8   decision is made in terms of treatment.

 

  9             As technology has advanced, eligibility

 

 10   criteria have moved beyond what they have been in

 

 11   the past, just being tumor histology and perhaps

 

 12   the staging of the patient.  As histologic and

 

 13   biologic characteristics of tumors are better

 

 14   defined and refined, we also are incorporating in

 

 15   many cases in pediatric oncology central input on

 

 16   the pathology and biology, such that central review

 

 17   of the patient's tumor pathology and diagnostic

 

 18   biology assays are used to improve the likelihood

 

 19   that a child receives the best available therapy

 

 20   for their specific tumor pathology and for their

 

 21   risk of relapse.

 

 22             This has been used in a variety of tumors

 

 23   in pediatric oncology in the recent past.  With

 

 24   rhabdomyosarcoma there is central review of

 

 25   alveolar versus embryonal rhabdomyosarcoma

 

                                                                75

 

  1   pathology that is used basically in real time so as

 

  2   to assure that the patient goes on the proper

 

  3   risk-stratified treatment regimen.  For

 

  4   neuroblastoma there are a variety of biologic

 

  5   characteristics that make amplification and other

 

  6   genetic changes that are characteristic to each

 

  7   tumor, and that is also looked at in real time.

 

  8   For Wilms tumor there has been a central review of

 

  9   that tumor histology for favorable histology versus

 

 10   focal or diffuse anaplasia that all distinguish

 

 11   patients for their appropriate trial, and there are

 

 12   a variety of genetic studies that are done, both

 

 13   centrally and locally, to establish the appropriate

 

 14   treatment for children with acute lymphoblastic

 

 15   leukemia, the most common diagnosis in childhood

 

 16   cancer.

 

 17             Phase I and pilot studies also have

 

 18   specific eligibility criteria.  In these cases, it

 

 19   may not necessarily be the case that you need to be

 

 20   concerned about the tumor histology so much,

 

 21   especially in Phase I where a child has already

 

 22   received treatment, but it is important to ensure

 

 23   that those patients who are enrolled in a trial

 

 24   have no other treatments that provide a reasonable

 

 25   potential for cure or substantial clinical benefit. 

 

                                                                76

 

  1   For patients who have newly diagnosed tumors but

 

  2   have a type of tumor that historically has a poor

 

  3   response to therapeutic interventions, we want to

 

  4   make sure that any sort of pilot treatment

 

  5   interventions that have been tried balance

 

  6   appropriately the benefits and likely risks in the

 

  7   child's prognosis.  So, before considering trial

 

  8   monitoring we consider that getting the right

 

  9   patient on the right trial is vital given the

 

 10   stratified approach we have to treatment in

 

 11   pediatric oncology.

 

 12             NCI supports a variety of investigator

 

 13   groups to do clinical trials in children with

 

 14   cancer.  The largest is the Children's Oncology

 

 15   Group, which pretty much every pediatric oncologist

 

 16   in North America is a member of.  That is the group

 

 17   that does the Phase III studies primarily as well

 

 18   as Phase II studies and pilot studies.  There is

 

 19   the COG Phase I Pilot Consortium that is a smaller

 

 20   group, about 20 institutions, that is assigned to

 

 21   do Phase I studies.  The Pediatric Brain Tumor

 

 22   Consortium I think is around 10 institutions as

 

 23   well.  Their focus is on newer therapies for brain

 

 24   tumors in children.  The new approaches to

 

 25   neuroblastoma therapy is a program project grant

 

                                                                77

 

  1   that NCI supports that is now 12 or 14 institutions

 

  2   I think, focused on early phase studies for

 

  3   children with neuroblastoma, high risk

 

  4   neuroblastoma.  There are also individual grants to

 

  5   investigators that may include clinical trial

 

  6   research.

 

  7             All these, because of the nature of

 

  8   pediatric oncology and the relative lack of number

 

  9   of patients, are usually multi-institutional.

 

 10   Given that they are multi-institutional, that

 

 11   brings on special responsibilities in terms of

 

 12   trying to conduct a trial at multiple sites

 

 13   simultaneously and trying to have all the

 

 14   investigators that are enrolling new patients and

 

 15   treating ongoing patients aware of what is going on

 

 16   with the trial.  So, the NCI has worked with these

 

 17   various groups that we support to facilitate this

 

 18   sort of intake of information and distribution of

 

 19   information.

 

 20             The investigators that are part of these

 

 21   various groups are committed to report toxicities,

 

 22   the regimen delivery and the ability to deliver the

 

 23   regimen as defined in the protocol and the response

 

 24   data in a timely fashion.  Some things such as

 

 25   remote data entry have been put in place now to

 

                                                                78

 

  1   help facilitate that.  There is a data center

 

  2   assigned with each of these groups that we support

 

  3   that is capable of readily receiving the data,

 

  4   analyzing the data and then reporting important

 

  5   data trends to the investigators, be it the study

 

  6   committee and perhaps beyond if necessary.  There

 

  7   is an operations office component.  They are able

 

  8   to communicate with investigators continuously

 

  9   throughout the clinical trial by email, by web

 

 10   site, by the phone, etc.  There is sort of this

 

 11   continuous back and forth going on between the

 

 12   investigators at the local institutions and a more

 

 13   centralized body that is helping to run the trial.

 

 14             In terms of monitoring, again it starts, I

 

 15   think just like enrollment, at the individual child

 

 16   level where there, is within the protocol, guidance

 

 17   provided to the local institutional clinicians as

 

 18   to what sort of laboratory results for

 

 19   tumor-related or treatment-related abnormalities

 

 20   need to be done and at what interval.  There are

 

 21   radiologic characterizations of the tumor and the

 

 22   consequent organ dysfunction that are also asked

 

 23   for in terms of the initial diagnosis of the child

 

 24   and then subsequently during their course of

 

 25   treatment.  Then there are interval evaluations to

 

                                                                79

 

  1   establish the tumor response to the treatment

 

  2   interventions that are being conducted during the

 

  3   study.

 

  4             The protocol--and we look for this at NCI

 

  5   when we review the protocols that come to us--must

 

  6   provide sort of a consistent and uniform approach

 

  7   to all these aspects of monitoring of the

 

  8   individual patient.  The frequency by which these

 

  9   studies are performed would be consistent with or

 

 10   greater than good clinical practice.  Because the

 

 11   children are on a clinical study, oftentimes they

 

 12   get more frequent monitoring of some of these

 

 13   aspects than they would if they received standard

 

 14   of care treatment off the protocol.  But, again, it

 

 15   depends on the intervention that is being

 

 16   undertaken and the specific tumor diagnosis under

 

 17   consideration.

 

 18             When you accumulate all this information,

 

 19   the monitoring and the clinical trial itself, that

 

 20   is where some of the infrastructure that NCI

 

 21   supports comes into play because, as I mentioned

 

 22   before, it is very important that patient data is

 

 23   submitted at protocol-defined intervals; that the

 

 24   data is accumulated, analyzed and then reported;

 

 25   and then that the significance of this data, be it

 

                                                                80

 

  1   the toxicity data or the effectiveness data, is

 

  2   interpreted so that appropriate patients are being

 

  3   accrued to the study; that treatment toxicity is

 

  4   acceptable and that there is some efficacy of the

 

  5   treatment interventions as defined in the protocol

 

  6   beforehand.

 

  7             There is some debate and discussion and

 

  8   variability in terms of who and how often this data

 

  9   that is accumulated and reported on is reviewed.

 

 10   Within NCI, we work with the guidelines established

 

 11   by NIH for data and safety monitoring and these

 

 12   requirements call for the oversight and monitoring

 

 13   of all human intervention studies to ensure the

 

 14   patient safety and the validity and integrity of

 

 15   the data itself for the study.  The monitoring in

 

 16   the study is to be done at sort of a level that is

 

 17   commensurate with the risks and size and complexity

 

 18   of the clinical trial.

 

 19             The oversight monitoring under Phase III

 

 20   clinical trials, which many of the pediatric

 

 21   oncology trials are, calls for the establishment of

 

 22   a DSMB.  The DSMB, according to NIH, is also

 

 23   appropriate for Phase I and Phase II clinical

 

 24   trials if the studies have such things as multiple

 

 25   clinical sites, are blinded or masked or employ

 

                                                                81

 

  1   particularly high-risk vulnerable patient

 

  2   populations.  In pediatric oncology we sort of hit

 

  3   throughout this so we call for sort of the default

 

  4   to be towards some sort of formalized monitoring

 

  5   committee for most of the studies that we do.

 

  6             The NCI, in response to NIH sort of

 

  7   formalizing its approach to data and safety

 

  8   monitoring, in the not too distant past has

 

  9   finished reviewing all the data and safety

 

 10   monitoring plans for the cancer centers that NCI

 

 11   supports across the country.  That was I think an

 

 12   education for both NCI as well as for the cancer

 

 13   centers, for them to really kind of fess up and

 

 14   look at what they actually do in terms of the

 

 15   monitoring; what goes on in their human subject

 

 16   clinical trials within their cancer centers.  But

 

 17   they all submitted them and they were all reviewed.

 

 18             Some of the key, essential elements for

 

 19   these monitoring plans that we had to consider, and

 

 20   that then subsequently have also been extended to

 

 21   some pediatric groups, are the monitoring and

 

 22   progress of the trials and safety of the

 

 23   participants; the plans for assuring compliance

 

 24   with adverse event reporting; and plans for

 

 25   assuring that data accuracy and protocol compliance

 

                                                                82

 

  1   are performed.

 

  2             As I mentioned, while in pediatric

 

  3   oncology basically we don't work from a cancer

 

  4   center model, we work more in a multi-institutional

 

  5   approach so it is a more distributed coverage in

 

  6   terms of who is performing the trials.

 

  7   Nevertheless, these particular essential elements

 

  8   were taken on by pretty much all the groups that we

 

  9   have that I mentioned earlier that NCI supports in

 

 10   one form or another, again, moving to the default

 

 11   of having some sort of more formalized data

 

 12   monitoring committees for all the trials.

 

 13             The composition of the DSMB and the

 

 14   various data monitoring committees may differ

 

 15   between the different groups that I mentioned that

 

 16   NCI supports for pediatric oncology but the goal is

 

 17   the same, and it is to have capable and informed

 

 18   observers be responsible for the oversight of the

 

 19   trial.  The reviewers are people that are outside

 

 20   of, and in addition to the study committee, and

 

 21   they evaluate the trial data at regular intervals

 

 22   to monitor the treatment toxicity and the

 

 23   effectiveness of the treatments that are being

 

 24   used.  Then, the review determines whether the

 

 25   continued accrual to the trial is safe and

 

                                                                83

 

  1   appropriate.  COG itself has two DSMBs, one for

 

  2   solid tumors and one for the leukemia and lymphoma

 

  3   studies, and they meet twice a year, each one of

 

  4   those DSMBs, to go over the studies.  Actually we

 

  5   go over pilot, Phase II and Phase III studies in

 

  6   those sessions.  The Phase I Consortia also has a

 

  7   DSMB that meets twice a year to go over all those

 

  8   Phase I studies.  In addition to the Phase I

 

  9   Consortia, the PBTC and the NANT, all of which have

 

 10   a DSMB type of component, have more frequent

 

 11   discussions with the groups that are beyond just

 

 12   the study investigator and any sort of data

 

 13   personnel or statistician directly involved.  They

 

 14   have a discussion of their studies sometimes on a

 

 15   weekly basis, sometimes on a monthly basis, and

 

 16   sometimes it also includes people from outside the

 

 17   group itself to overlook what is going on with

 

 18   their particular studies.

 

 19             In terms of compliance with adverse event

 

 20   reporting, another one of the essential elements

 

 21   that NCI has, NCI-funded studies use the adverse

 

 22   event expedited reporting system, or the AdEERS

 

 23   system to report toxicities.  This is a

 

 24   computerized system that is available now to all

 

 25   the funded groups with which they can fairly easily

 

                                                                84

 

  1   report adverse events that occur during their

 

  2   clinical trials.  That data can then be accumulated

 

  3   easily within their group, but also important

 

  4   things can be sent off to the FDA or to drug

 

  5   sponsors or the NCI as appropriate, especially for

 

  6   studies that involve IND agents.

 

  7             Then, it is the institutional principal

 

  8   investigator that is ultimately responsible to

 

  9   assure that the AEs are reported in a timely

 

 10   manner.  Whenever we review the cancer center

 

 11   approaches, they list out that sort of the CRA

 

 12   should submit this and then there is a nurse

 

 13   practitioner or someone that is behind the CR to

 

 14   make sure it gets submitted, and at some interval

 

 15   the principal investigator locally is responsible

 

 16   to make sure that all the AEs that may have

 

 17   occurred had been properly reported.

 

 18             Finally, for assuring data accuracy and

 

 19   protocol compliance, the cooperative groups and

 

 20   these consortia practice ongoing quality control

 

 21   and interval quality assessments such as by using

 

 22   institutional audits.  This has been something that

 

 23   has been ongoing throughout the creation of each of

 

 24   these groups.

 

 25             In summary, NCI has worked to establish a

 

                                                                85

 

  1   framework to allow appropriate monitoring and

 

  2   oversight of pediatric oncology clinical trials.

 

  3   To address some of the issues that Steven had

 

  4   brought up before in terms of the general

 

  5   parameters that we look at, we first want to make

 

  6   sure that the enrollment of patients is appropriate

 

  7   to the diagnosis and risk of relapse for the

 

  8   patient or the availability of standard treatments

 

  9   for recurrent and relapsed disease, and that

 

 10   laboratory and radiologic monitoring for toxicity

 

 11   and response to treatments is established within

 

 12   the protocol before any patients are accrued.

 

 13             The frequency of monitoring would be equal

 

 14   to or greater than standard of care for the

 

 15   individual patient that is enrolled on a clinical

 

 16   study, and there would be continuous protocol

 

 17   monitoring by the study committee because they

 

 18   receive this data on a daily basis.  There would be

 

 19   interval protocol monitoring on a monthly to

 

 20   biannual basis, depending on the risk and specifics

 

 21   of the trial, by a group outside of the study

 

 22   committee itself.

 

 23             Who does the monitoring?  The daily

 

 24   monitoring is by the study committee itself.  The

 

 25   interval monitoring usually involves concentrations

 

                                                                86

 

  1   and statisticians that are not directly involved in

 

  2   the trial.

 

  3             When is a data monitoring committee

 

  4   needed?  For Phase III studies you need a DSMB.

 

  5   For multi-institutional trials you need to have a

 

  6   monitoring committee for high-risk populations.

 

  7   You need to have a monitoring committee for complex

 

  8   treatment.  For studies with early stopping rules,

 

  9   which many pediatric studies have, you have to have

 

 10   a monitoring committee.  With conflicts of

 

 11   interest, which may not be as much of a case in

 

 12   pediatrics as it might be in medical oncology, you

 

 13   need to have a monitoring committee.

 

 14             I think that with pediatric oncology

 

 15   trials we hit many of the points that are brought

 

 16   up by various agencies of situations where a

 

 17   monitoring committee is required so that virtually

 

 18   always in pediatric oncology some sort of

 

 19   monitoring committee is involved in the oversight

 

 20   of the practices of the group, as well as the

 

 21   conduct of individual clinical trials.  Thank you.

 

 22             DR. SANTANA:  Thanks, Barry.  Before I

 

 23   stand up to give the last presentation of the

 

 24   morning, we have an opportunity for an open public

 

 25   hearing.  So, if there is anybody in the audience

 

                                                                87

 

  1   that wishes to address the committee, this is the

 

  2   opportunity to do so.  I would ask that if you are

 

  3   going to do that you come to the front of the room

 

  4   to the podium and identify yourself by name and

 

  5   affiliation.

 

  6                       Open Public Hearing

 

  7             MR. RAKOFF:  Wayne Rakoff, Johnson &

 

  8   Johnson.  Just a quick question, that came up this

 

  9   morning that I would like to hear discussed during

 

 10   the discussion, is with regard to the FDA guidance

 

 11   on data reduction in oncology trials.  It would be

 

 12   important to us to know if there are any variances

 

 13   in that with regard to pediatric studies.

 

 14             DR. SANTANA:  Steve or Rick, do you want

 

 15   to address that now or do you want to address it

 

 16   during the discussion period?

 

 17             DR. HIRSCHFELD:  We can address it in a

 

 18   little more detail but, in brief, that is a global

 

 19   commentary and there isn't a specific pediatric

 

 20   component to it.  I think that is a good suggestion

 

 21   that maybe we should consider in the future, a

 

 22   pediatric specific component.

 

 23             DR. SANTANA:  Any other comments from the

 

 24   audience?

 

 25             [No response]

 

                                                                88

 

  1                Monitoring Procedures at a Private

 

  2                       Children's Hospital

 

  3             DR. SANTANA:  First of all, I want to

 

  4   thank Steve, Richard and the rest of the FDA for

 

  5   always bringing the pediatric oncologists to set

 

  6   examples in these initiatives.  I am personally

 

  7   very appreciative of all the efforts that we have

 

  8   had on behalf of the issues that we deal with in

 

  9   pediatric oncology.

 

 10             My task this morning, as I was charged to

 

 11   do, is to bring a perspective from a private

 

 12   institution with the caveat that St. Jude really is

 

 13   an NCI cancer designated center so a lot of what we

 

 14   do in terms of our own monitoring is reflective of

 

 15   what we have to do to comply with the NCI

 

 16   regulations.

 

 17             What I would like to do over the next 20

 

 18   minutes or 25 minutes or so is talk to you about

 

 19   two issues.  One is how we set forth monitoring of

 

 20   our St. Jude studies--not the cooperative group

 

 21   studies for which we still have to comply with COG,

 

 22   but our own intra-institutional studies that follow

 

 23   a parallel system to the NCI monitoring plan, and

 

 24   what that monitoring plan involves and what

 

 25   parameters we have designated for monitoring. 

 

                                                                89

 

  1   Then, a bigger part of my talk will be on a project

 

  2   that Don Workman and I worked on in terms of trying

 

  3   to handle adverse event reporting within the

 

  4   institution and tried to develop an interactive

 

  5   web-based model to try to get a handle on that.

 

  6             With that, I will go ahead and get

 

  7   started.  As Barry has already said, monitoring of

 

  8   trials is really an ongoing, continuous review of

 

  9   the conduct of the trial.  For the purpose of

 

 10   distinction, I will make the note that to me

 

 11   monitoring occurs while the study is ongoing.

 

 12   Whereas a lot of people use the word auditing, to

 

 13   me auditing is a post facto thing that happens

 

 14   after the study has been completed.  Then you go

 

 15   back and see if the study was conducted the way it

 

 16   was supposed to be; if the data is good enough; if

 

 17   there is quality in the data; and if there have

 

 18   been any other issues that occurred during that

 

 19   post facto process.  So, to me, monitoring occurs

 

 20   real time whereas auditing occurs after the study

 

 21   has been completed.

 

 22             Monitoring is really a shared

 

 23   responsibility of many individuals.  We always talk

 

 24   about monitoring being the responsibility of maybe

 

 25   one particular group but at St. Jude we have the

 

                                                                90

 

  1   notion that this is really the responsibility of

 

  2   the research team.  We always talk about the

 

  3   principal investigator but it is really the

 

  4   research team.  The research team has many

 

  5   components to it of which, hopefully, the principal

 

  6   investigator is the lead person but there are

 

  7   research nurses, there are CRAs, there are other

 

  8   members of the study team who also have

 

  9   responsibility for this process.

 

 10             Institutional officials have a major role

 

 11   in this, not only in terms or providing

 

 12   infrastructure resources to conduct some of this

 

 13   monitoring, but also to set a culture and example

 

 14   that is transparent to make sure that things occur

 

 15   very openly and that everybody is knowledgeable

 

 16   about what is happening.  Then, the oversight

 

 17   committee--you heard a little bit about DSMBs which

 

 18   I won't talk about and IRBs and other committees

 

 19   that may be involved in this process.

 

 20             Eric had a little figure this morning of a

 

 21   triangle.  I didn't know he had a triangle so I

 

 22   brought a triangle too, but my triangle is a little

 

 23   bit different.  It makes a different point.  The

 

 24   point of this triangle is that in the center of the

 

 25   process are the participant in the research but

 

                                                                91

 

  1   there are many other people involved in this whole

 

  2   process in which, as I mentioned to you earlier,

 

  3   the partnership includes the investigator, the

 

  4   research team, the IRB, other oversight committees

 

  5   and then institutional officials.  So, I view this

 

  6   more as a partnership, not just the responsibility

 

  7   of one individual.

 

  8             One of the things I want to cover is point

 

  9   number one and point number three on this slide,

 

 10   which is how can we systematically approach some of

 

 11   these problems in terms of monitoring and adverse

 

 12   event reporting.

 

 13             So, I think the first step whenever you

 

 14   deal with a promise to define a problem in this

 

 15   case is what needs to be monitored and what needs

 

 16   to be reported.  I think that is a good point to

 

 17   start and I will talk about that in a minute; then,

 

 18   dividing the role, the different committees that

 

 19   provide some of this oversight and I really won't

 

 20   go into detail on that although I could during the

 

 21   discussion if anybody has any questions; and,

 

 22   lastly, developing an infrastructure to allow this

 

 23   to happen so that the reporting occurs, that there

 

 24   is a process of evaluating the reports, and then a

 

 25   process of acting in a timely manner when there are

 

                                                                92

 

  1   concerns.  So, that will be the latter part of my

 

  2   talk.

 

  3             As I mentioned to you, we are an NCI

 

  4   cancer designated center so we also had to comply

 

  5   and submit an institutional data safety monitoring

 

  6   plan to the NCI a few years back that was reviewed,

 

  7   approved, etc., etc., and now we provide our

 

  8   monitoring under the umbrella of what that plan

 

  9   says.

 

 10             So, the first thing was to define what

 

 11   elements we were going to monitor.  So, we have

 

 12   kind of followed the parallel system that the NCI

 

 13   designated in the clinical data update system of

 

 14   what data should be collected.  We look at patient

 

 15   specific data, the demographics, date of birth,

 

 16   gender, those things that we have to collect; the

 

 17   date of entry into the study; the treatment status,

 

 18   if the patient has been previously treated, on what

 

 19   protocols and what therapy the patient was on; and,

 

 20   if they were off therapy, for what reasons.  All

 

 21   that gets captured as part of the monitoring of the

 

 22   patient on the study.

 

 23             Then, there are subgroup data elements

 

 24   that are also captured.  Barry mentioned, very

 

 25   appropriately in his talk, the issue of eligibility

 

                                                                93

 

  1   and determining that the right patients go on the

 

  2   right studies.  One of the things we have done at

 

  3   St. Jude in the last ten years is we have

 

  4   established a separate office, which is called the

 

  5   protocol office which is actually an office that

 

  6   provides the infrastructure to help investigators

 

  7   deal with many of these issues.  The protocol

 

  8   office, obviously, is manned by a group of people

 

  9   and one of the responsibilities, for example, is

 

 10   that when an investigator enrolls a patient on a

 

 11   study we have to fill out electronically an

 

 12   eligibility check list.  The eligibility check list

 

 13   gets faxed to that office and a patient-specific

 

 14   consent is generated for that patient on that

 

 15   study.  So, right at the beginning there are some

 

 16   checks and balances in terms of the eligibility of

 

 17   the patient so that the right patient is put on the

 

 18   right study and the correct consent is used for

 

 19   that patient.  So, that is an ongoing process that

 

 20   occurs early on during the trial and the patient

 

 21   enrollment of the trial.

 

 22             Once the patient receives the therapy,

 

 23   they monitor the cycle or the course of therapy.

 

 24   If is a Phase I study, what dose level the patient

 

 25   is currently being treated with; the start date;

 

                                                                94

 

  1   some other parameters like BSA and weight.  They

 

  2   monitor, particularly in Phase I studies, the

 

  3   agent; the dose of the agent; if there have been

 

  4   any modifications, why there have been

 

  5   modifications.  We will talk a little bit about

 

  6   adverse event reporting later on.  Then, as part of

 

  7   the monitoring during certain periods of the trial,

 

  8   the patients will be monitored in terms of response

 

  9   because the trials will have stopping rules based

 

 10   on response, not only in terms of toxicity but also

 

 11   in terms of response so a Phase II trial that has

 

 12   some response built-in stopping rules will be

 

 13   stopped at the right point once the monitoring is

 

 14   occurring in terms of the response that has been

 

 15   achieved.

 

 16             I tried to summarize this in two or three

 

 17   slides.  This is kind of how we do it at St. Jude

 

 18   in terms of our own institutional Phase I/Phase II.

 

 19   We don't do many Phase III but we do have an

 

 20   auditing plan for Phase III studies and for some

 

 21   studies in which we hold the IND.

 

 22             So, for Phase I studies the central

 

 23   elements in terms of demographics, eligibility and

 

 24   informed consent, that is monitored continuously.

 

 25   It is monitored continuously because I told you

 

                                                                95

 

  1   that there is a check at the beginning in terms of

 

  2   eligibility and in terms of informed consent that

 

  3   occurs in real time when the patient gets

 

  4   registered.  So, that is done continuously as the

 

  5   patients go on a study in a Phase I study.

 

  6             The protocol office also is monitoring the

 

  7   study in terms of the data elements for the study

 

  8   so there are templates very similar to the RDE

 

  9   system that is developed by COG, templates of data

 

 10   capture forms.  Those data capture forms are

 

 11   electronic and the monitor on a monthly basis that

 

 12   he or she is assigned will go through those and

 

 13   will see if there is data that is missing.  If

 

 14   there is data that is missing, a report is

 

 15   generated to the principal investigator that data

 

 16   is missing on a monthly basis.  So, it is a good

 

 17   system in terms that it keeps the research team

 

 18   kind of continuously on top of making sure the data

 

 19   is being collected.

 

 20             On a quarterly basis for a Phase I study

 

 21   there is a report that is generated.  I will show

 

 22   you in a minute where the reports go but, in a

 

 23   nutshell, it goes, obviously, to the principal

 

 24   investigator and to the research team, and then it

 

 25   goes to the subcommittee of the scientific review

 

                                                                96

 

  1   committee that also oversees monitoring to make

 

  2   sure that they are separate from the protocol

 

  3   office and from the investigator looking at this

 

  4   data.

 

  5             Then, for every Phase I study that we are

 

  6   the primary sponsor of at St. Jude, the first three

 

  7   patients enrolled in the study are monitored.

 

  8   Then, once the first three patients are monitored,

 

  9   one additional patient per dose level is monitored

 

 10   in real time.  The idea of doing the first three

 

 11   patients is that in many studies usually within the

 

 12   first three patients you know if your systems are

 

 13   in the right checks and balances so that you want

 

 14   to monitor those first three patients very acutely

 

 15   so if there is a problem with the system, with the

 

 16   templates, with potentially things not going right,

 

 17   you can pick it up very quickly and make the right

 

 18   adjustment so that for the subsequent dose levels,

 

 19   if you monitor one patient in real time, you should

 

 20   have resolved all of that.

 

 21             We do a lot of Phase II studies at St.

 

 22   Jude and we also do the eligibility, essential

 

 23   elements and consents as outlined here.  We also do

 

 24   missing data reports on a quarterly basis.

 

 25   Obviously, in Phase II, just like in Phase I, you

 

                                                                97

 

  1   are interested in adverse events and those are

 

  2   reported quarterly.  Then, on a semiannual basis

 

  3   the monitors will verify the coding of response so

 

  4   that the studies can be stopped if the response

 

  5   criteria for stopping rules have been met.  There

 

  6   are reports semiannually or more frequently or less

 

  7   frequently, as defined by the protocol, in terms of

 

  8   the individual monitoring plan that the protocol

 

  9   may have.

 

 10             In Phase II we always monitor the first

 

 11   two patients plus at least--and the clever word

 

 12   here is "at least" ten percent of the total

 

 13   patients that are being accrued.  It could be

 

 14   greater than ten percent.  It depends obviously on

 

 15   the resources that you have available and the

 

 16   workload that the specific monitor may have but at

 

 17   a minimum ten percent of the patients on any Phase

 

 18   II study at any given time should be under active

 

 19   monitoring.

 

 20             We don't do many Phase III studies at St.

 

 21   Jude but we do have a marching plan in the event

 

 22   that there is a Phase III study and it parallels

 

 23   the Phase II monitoring plan, with the exception

 

 24   that there may be other primary objectives in the

 

 25   Phase III trials that also require some monitoring.

 

                                                                98

 

  1             St. Jude holds INDs or IDEs for a few

 

  2   products so under those circumstances, they could

 

  3   be Phase I or Phase II trials or whatever, but

 

  4   separately from those, if there is a particular IND

 

  5   or IDE for which St. Jude is the "sponsor" then

 

  6   there is a specific monitoring plan that is

 

  7   assigned to that study, and it will depend on the

 

  8   risk, what is known about the IND drug, what is

 

  9   known about the device, etc., etc., and may be more

 

 10   strict but at least it will be just like Phase I or

 

 11   Phase II studies I described to you before.

 

 12   Usually, under some circumstances like some novel

 

 13   therapy, it may be a little bit stricter in that

 

 14   the studies are being monitored a little bit more

 

 15   aggressively.

 

 16             So, this is kind of in a nutshell how we

 

 17   kind of agree with the NCI in our data safety

 

 18   monitoring plan and how we would monitor our

 

 19   studies.  Having said that, there is also auditing

 

 20   that occurs.  So, there is a different auditing

 

 21   plan that I am going to give a lot of detail about,

 

 22   but for most auditing plans the monitors, once the

 

 23   study is done, will make sure that at least 20

 

 24   percent of the patients have had a full audit of

 

 25   their records.  But that is after the study is done

 

                                                                99

 

  1   and that occurs over a long period of time.  It is

 

  2   not as active as the actual monitoring which is

 

  3   occurring in real time.

 

  4             I want to switch now and talk a little bit

 

  5   about the issue of adverse event reporting which

 

  6   has to do with monitoring and safety.  We, at St.

 

  7   Jude, also have struggled with this issue and we

 

  8   struggle because there are a lot of problems in

 

  9   reporting.  There tends to be a lot of

 

 10   over-reporting.  That is, anticipated adverse

 

 11   events that are known in the investigator's

 

 12   brochure or known from other clinical trials are

 

 13   being reported on a continuous basis and that

 

 14   creates a big backlog of data that is important but

 

 15   not important in real time in terms of monitoring.

 

 16             As you all know, there is increased

 

 17   research in new drugs and biologics.  There is more

 

 18   oversight and scrutiny by federal agencies.  Just

 

 19   like in many other places, we tend to get

 

 20   saturation effects.  There comes a point where you

 

 21   see so many reports that it doesn't ring a bell; it

 

 22   doesn't ring any whistles or anything like that.

 

 23   So, we have to be careful that we don't over-report

 

 24   because then it gets us into the saturation effect

 

 25   and we don't react appropriately when there are red

 

                                                               100

 

  1   flags that we should be paying attention to.

 

  2             But one of the problems we have at St.

 

  3   Jude, which is very common for pediatric

 

  4   institutions, is that there are no denominators for

 

  5   how to make any sense of this; what constitutes a

 

  6   red flag?  Where do you cut the line to say this is

 

  7   important or this is not important?  There is no

 

  8   normative data for each of the populations that we

 

  9   have to deal with for Phase I studies, for Phase II

 

 10   studies and for the studies I mentioned to you.

 

 11   So, trying to approach this problem, we have tried

 

 12   to deal with this I think in a prospective way.

 

 13             In terms of review, there are a lot of

 

 14   external events that we get from study sponsors.

 

 15   If there happens to be a drug that we are doing a

 

 16   study with but the drug is being used in adult

 

 17   studies or in other institutions, you know, the

 

 18   sponsors package a lot AEs and send them to you and

 

 19   we have to deal with those too.  The problem with

 

 20   those is that sometimes the information is very

 

 21   sketchy and there is no opportunity for

 

 22   clarifications or for questions so that then you

 

 23   can put that in the context of your own experience

 

 24   with your own patients at your own institution.

 

 25             The other thing is that the IRB is not a

 

                                                               101

 

  1   DSMB.  A DSMB has a very specific role; the IRB has

 

  2   to deal with a lot of other issues.  They have to

 

  3   deal with adverse events and they should be looking

 

  4   at them and they should be judging them, but it is

 

  5   clearly in the context of the whole package of the

 

  6   research, whereas the DSMB has very specific roles

 

  7   and responsibilities.

 

  8             The IRB is not the FDA who holds the IND

 

  9   file for the drug and knows everything.  So, the

 

 10   IRB over here is getting little pieces of

 

 11   information and trying to make sense out of it in a

 

 12   more global sense.  Then, the IRB also needs to

 

 13   rely on the local investigators to interpret the

 

 14   meaning of the adverse events that they are

 

 15   receiving from the outside, from the sponsors,

 

 16   because clearly the IRB doesn't have the expertise

 

 17   or the knowledge to put that in contextual features

 

 18   in terms of the study as it is being conducted at

 

 19   other institutions.

 

 20             So, at St. Jude we decided to approach

 

 21   this problem first by doing quality improvement

 

 22   projects, trying to figure out where the problems

 

 23   were and where we could attach the problems.  One

 

 24   of the first issues that we addressed is that at

 

 25   the beginning the PI or the research team needs to

 

                                                               102

 

  1   report and categorize the events, but there was no

 

  2   systematic way of doing that.  I mean, it was being

 

  3   done in paper form; there were different versions

 

  4   of that paper form.

 

  5             One of the things that Don Workman and I

 

  6   recognized is that at least if at the beginning we

 

  7   could make this a standardized way and force

 

  8   everybody to do it the same way, then five, ten

 

  9   years later we actually would have a system in

 

 10   place that would provide a lot of the normative

 

 11   data that we would need in order then to do some

 

 12   process improvement.

 

 13             So, the first thing that we did is to

 

 14   create this electronic submission that I will

 

 15   describe to you in a few minutes.  This electronic

 

 16   submission is pretty neat I think, to use words of

 

 17   my nephew--it is pretty neat because it allows you

 

 18   then to disseminate that information very quickly

 

 19   to all the key players in the field and then they

 

 20   can do their own assessment the same time that the

 

 21   IRB is doing their assessment.  So, the IRB will

 

 22   get a copy of this electronic adverse event and the

 

 23   IRB will do their own assessment of the adverse

 

 24   event and certainly give feedback and follow-up to

 

 25   the investigator.  At the same time that it goes to

 

                                                               103

 

  1   the IRB, it goes to our office of regulatory

 

  2   affairs which is also charged with making sure that

 

  3   agencies that have to be notified about these

 

  4   adverse events are also notified.  So, it kind of

 

  5   takes the IRB and the investigator away from that

 

  6   responsibility of having do to that paperwork but

 

  7   it goes to a central office that then now deals

 

  8   with all the external agencies that have to look at

 

  9   this data.

 

 10             Internally, it goes in a different

 

 11   direction.  It goes to the vice president of

 

 12   clinical trials for internal reporting and internal

 

 13   processing so that the St. Jude DSMB or what we

 

 14   call our scientific review council which is called

 

 15   the CPSRMC, the clinical protocol scientific review

 

 16   monitoring committee, is really the scientific

 

 17   council which also has a function in terms of the

 

 18   cancer center doing monitoring.  They also get a

 

 19   copy of the report and then they deal with it

 

 20   internally and then they can give also feedback to

 

 21   the principal investigator.

 

 22             Don and I were very concerned with the

 

 23   first step in this process to try and make it

 

 24   uniform and to try to make it normative so that we

 

 25   could then create a system that, hopefully, would

 

                                                               104

 

  1   help us in retrospect.  So, we started this about

 

  2   18 months ago.  The first thing we did is we said

 

  3   let's create a form that is standardized.  We can

 

  4   then make sure that people understand what is

 

  5   important in that form before we convert it into an

 

  6   electronic format.  Then we were able, as we

 

  7   designed the form, to start thinking prospectively

 

  8   of how that same data could be captured

 

  9   electronically.

 

 10             Then we developed a flow diagram as a

 

 11   quality improvement project of where this web-based

 

 12   report could go, which is a little bit of what I

 

 13   just showed you.  We had to deal with some issues

 

 14   of security access and then we also had to deal

 

 15   with some issues of electronic signature that we

 

 16   eventually resolved.

 

 17             One of the key features of this, which is

 

 18   a recurrent problem in adverse event reporting, is

 

 19   that there are databases and the databases don't

 

 20   talk to each other.  So, one of the key features

 

 21   that we wanted to cover in this was to make sure

 

 22   that this adverse event electronic reporter was

 

 23   talking to the other databases in the hospital and

 

 24   was capturing information from the protocol office

 

 25   in terms of the protocol that the patient was

 

                                                               105

 

  1   registered on and the additional protocols was that

 

  2   the patient was registered on because there could

 

  3   be some cross-talk between adverse events on

 

  4   different protocols or different PIs.  I will show

 

  5   you an example at the end.

 

  6             We also wanted to make this user friendly

 

  7   and make sure that anybody who is part of the

 

  8   research team could do this at any place in the

 

  9   hospital.  Through a security pass they could

 

 10   access this web site and could potentially feed in

 

 11   the information in a very quick manner, without

 

 12   having to go to a dark office somewhere and grab

 

 13   papers and try to do it.  So, there were some

 

 14   security access issues that got resolved but it was

 

 15   made available to anybody on the research team

 

 16   electronically.

 

 17             We then tried to address the issue of

 

 18   internal reporting, that is studies in which

 

 19   adverse events are occurring in our patients at our

 

 20   institution versus the information of adverse

 

 21   events that are occurring at other sites that are

 

 22   being fed into our protocols in terms of the

 

 23   cooperative group studies, and so on and so forth.

 

 24   So, one of the things that we had to address is how

 

 25   we could link protocols so that the information

 

                                                               106

 

  1   could be identified very easily.  If a patient was

 

  2   registered on one protocol and the adverse event

 

  3   occurred on that protocol, we wanted to know what

 

  4   additional studies that patient was enrolled on so

 

  5   that when the IRB or the subcommittees reviewed

 

  6   this they could begin to get trends if there were

 

  7   complementary adverse events that were occurring

 

  8   from complementary studies and there could have

 

  9   been a red flag there that we needed to address.

 

 10             In addition, we could share the

 

 11   information with the PIs of the other studies

 

 12   because they also have to be kept in the loop in

 

 13   terms of what is happening to patients that

 

 14   potentially may also be enrolled in their own

 

 15   studies concomitantly, for example therapeutic

 

 16   versus non-therapeutic studies.

 

 17             Then, for external reports we wanted the

 

 18   investigators to help us sort that out because we

 

 19   couldn't sort it out.  So, the investigators had to

 

 20   invest some time at the beginning sorting out

 

 21   external reports before they submitted them to us

 

 22   so that they would be more meaningful to us.

 

 23             Then, the functional outcomes would be

 

 24   that there would be real-time reporting and that

 

 25   the IRB would acknowledge that through some

 

                                                               107

 

  1   electronic time stamping mechanism.  There are

 

  2   forced choices so that everybody has to do it the

 

  3   same way; no incomplete data submissions so we

 

  4   wouldn't have to address the issue of going back

 

  5   and asking for more clarification and more

 

  6   questions; easy access so it would be friendly;

 

  7   ability to generate single incident reports;

 

  8   ability to generate reports in a given time period.

 

  9   If you were noting a trend that something was

 

 10   occurring in a particular study over some period of

 

 11   time, you could capture that and, as you will see

 

 12   in the end, provide cumulative data that you could

 

 13   sort out to look at trends that potentially could

 

 14   be occurring.  Quicker reporting times; ability for

 

 15   the IRB office to generate reports based on

 

 16   protocols; specific events across subjects, across

 

 17   protocols to give us some functionality at the IRB

 

 18   level to look at the data in different ways;

 

 19   generate internal denominators of trends that we

 

 20   wanted to look at; use standardized NCI toxicity

 

 21   tables for the oncology trials; and be able to

 

 22   record the IRB actions and updates from

 

 23   investigators onto previous reports.  So, it wasn't

 

 24   a dead system.  It was a system that the

 

 25   investigator could go back and add more information

 

                                                               108

 

  1   or, when the IRB reviewed it, could add more

 

  2   information so it became a living document as the

 

  3   report was being done.

 

  4             Let me give you an example of how this

 

  5   works.  I couldn't get it electronically.  It was

 

  6   going to cost me money to be able to do this

 

  7   electronically so I did some snapshots of what it

 

  8   looks like.

 

  9             So, this page is accessible to anybody who

 

 10   is identified at St. Jude as a principal

 

 11   investigator or a member of a research team.  So,

 

 12   if you are listed on the protocol as the nurse for

 

 13   that study, as the statistician for that study, as

 

 14   a pharmacist for that study, automatically you get

 

 15   access to this through a user ID and your own

 

 16   password.  So, it is available to anybody who is

 

 17   part of the research team.

 

 18             This is how you log in.  Here I logged in

 

 19   and it says, "welcome, Victor Santana."  Then it

 

 20   gives a listing of all the events that have

 

 21   accumulated during a particular period of time.  It

 

 22   gives the event ID which is an internal working

 

 23   number.  It gives the event date.  It gives an

 

 24   identifier that I have erased here for a particular

 

 25   patient.  It is usually a numerical number.  If it

 

                                                               109

 

  1   is an external event, then there is a way to code

 

  2   that to an external number.  Sometimes you get an

 

  3   event from a sponsor and it is coded ABXY235, well,

 

  4   there is a way that you can code that the same way

 

  5   here so you can track it and use the same codifier

 

  6   if you ever have to go back to the data.

 

  7             The status tells me, as an investigator,

 

  8   whether I have reviewed this or not.  So, when I

 

  9   copied this the other day I only had one adverse

 

 10   event that I had yet to review that somebody sent

 

 11   to me for comment.  Then, it tells me the date that

 

 12   the event was reviewed by me or that I modified it

 

 13   or I did anything to it.

 

 14             Very quickly, it goes through a couple of

 

 15   screens that provide some general information.  It

 

 16   tells you whether it is a St. Jude patient or not

 

 17   because if it is not, it throws you in a different

 

 18   direction in terms of the data that you need to

 

 19   capture because, clearly, the data is being

 

 20   captured for external adverse events a little bit

 

 21   differently than it is for internal.  There is some

 

 22   information here in terms of the patient.

 

 23             Then, it begins to do its own internal

 

 24   processing once it identifies the patient.  It

 

 25   tells us, as you see at the top of the screen, all

 

                                                               110

 

  1   the protocols that this patient is registered on.

 

  2   So, it goes back and talks to the data warehouse.

 

  3   If this patient is enrolled on ten studies, it will

 

  4   pull and identify all those ten studies.  Then it

 

  5   will ask me, as the person putting in the

 

  6   information, under what study am I following this

 

  7   report.  So, it identifies primarily the study and

 

  8   the adverse event, but it also tells me all the

 

  9   other studies the patient is on, and this is

 

 10   critical because this report will go to the PIs of

 

 11   all those other studies too.  You will see it at

 

 12   the end for their comments.  So, it provides a

 

 13   little bit of a cross-talk among studies.

 

 14             Then, it clearly identifies the type of

 

 15   adverse event that is being reported.  You have all

 

 16   seen this in different variations.  For adverse

 

 17   events that require a CTC code it takes you to the

 

 18   CTC code so there is a link too so you don't have

 

 19   to scramble through 50 books looking for those

 

 20   codes but automatically it links you to those

 

 21   codes.  Then, it allows you to put the descriptor,

 

 22   etc., etc.  So, it is all being captured in a

 

 23   uniform language.

 

 24             Then it goes to a page that allows the

 

 25   person who is submitting the information to do some

 

                                                               111

 

  1   attribution on the adverse event.  It is a click

 

  2   system but it reminds people, because we all tend

 

  3   to forget, what each one of those words means.  So,

 

  4   it reminds me that I need to read when something is

 

  5   serious; when something is unexpected.  It defines

 

  6   it very clearly because there are always a lot of

 

  7   questions from members of the research team what

 

  8   constitutes something that is unexpected versus

 

  9   expected.  Well, there it is.  It is, hopefully,

 

 10   black and white and then you select, based on your

 

 11   interpretation.  It allows you to do one selection

 

 12   across lines horizontally for each one of those.

 

 13             Then, there is a page that allows you to

 

 14   provide more information.  One of the problems

 

 15   always with electronic information is that

 

 16   sometimes you can't capture everything in a unique

 

 17   format.  So, there is a page that allows you to do

 

 18   a little more narrative form of how this all

 

 19   happened, and so on and so forth, so it can give

 

 20   you some additional data that you can comment on.

 

 21             Then it asks you do you think, based on

 

 22   your interpretation of what has happened with the

 

 23   adverse event, that there is a follow-up that is

 

 24   needed.  If you say there is a follow-up needed,

 

 25   then it links back to a reminder within 30 days

 

                                                               112

 

  1   that you owe us a follow-up.  The IRB reviews it

 

  2   and they also communicate directly.  But if you

 

  3   think you have enough information and you want to

 

  4   submit a follow-up, within 30 days you will get a

 

  5   reminder that you owe us a follow-up.

 

  6             Then it tells you something about what

 

  7   happened to the patient based on that adverse

 

  8   event.  Then it asks the investigator or the

 

  9   research team to make some judgments based on the

 

 10   information that they have on that particular

 

 11   adverse event, and in terms of what they know is

 

 12   going on in the study does this alter the

 

 13   risk/benefit ratio for the other participants.

 

 14   Does this require modifications to the protocol or

 

 15   to the consent?  And, does this provide additional

 

 16   information that we should be sharing with other

 

 17   people that are participating in the study?  So, we

 

 18   ask the investigator to specifically address these

 

 19   issues with each adverse event.

 

 20             This is an example of a summary page.  All

 

 21   that data is generated in the end into a summary

 

 22   page.  Obviously, I have whited out a lot of stuff.

 

 23   There is a doctor that is called "Dr. Teddy Bear."

 

 24   That is a famous doctor at St. Jude that we always

 

 25   use whenever we do electronic examples of things. 

 

                                                               113

 

  1   But it gives you a nice summary of who is doing

 

  2   this; who reported it; the protocol which was

 

  3   reported; the PI of that study; the date it was

 

  4   reported; when the adverse event started.  It will

 

  5   list all the studies, based on that warehouse

 

  6   capture of data, that the patient was on.  It will

 

  7   quickly generate all that data into specifically

 

  8   designated toxicities that were reported as part of

 

  9   the adverse event.  The attribution and nature that

 

 10   you selected gets summarized; additional medical

 

 11   history; treatment prognosis; patient outcome.

 

 12             Then it tells us at the end--this all goes

 

 13   to the IRB--it tells us at the end how the

 

 14   principal investigator judged this in terms of his

 

 15   own interpretation, that it doesn't alter the

 

 16   risk/benefit ratio; does not require modification,

 

 17   and so on and so forth.

 

 18             So, it goes electronically--only focusing

 

 19   on the IRB part of this, it goes electronically to

 

 20   the IRB and there is a designated person in the IRB

 

 21   office who will certify that he or she has received

 

 22   this report, and will certify it electronically

 

 23   down here with the date.  Then it allows, at the

 

 24   end, to add additional information when the IRB

 

 25   actually reviewed it.  So, the IRB will come back

 

                                                               114

 

  1   at the end of the meeting and put in there the date

 

  2   that it was reviewed by the IRB so it provides a

 

  3   tracking record of when the IRB looked at it.

 

  4             Another very neat thing I think, and I

 

  5   like to use that word, with this project was that

 

  6   it allowed cross-communication among investigators.

 

  7   In that example I gave you, the message that there

 

  8   was an adverse event reported in August, '99 will

 

  9   also go to all these other studies that that

 

 10   patient was enrolled on.  So, the PI of the SD/01

 

 11   protocol will also get the message and will get the

 

 12   summary report, and the PI of that study has to pay

 

 13   attention to that report and then make a decision

 

 14   whether he or she thinks it may or may not be

 

 15   related to his study too because there could be

 

 16   complementary toxicities and they are the only ones

 

 17   who are going to know that, not the IRB unless it

 

 18   gets reported through a different mechanism.

 

 19             So, it forces all the PIs of all the

 

 20   studies that the patient is enrolled on to also

 

 21   critically review the adverse event and make some

 

 22   judgment about whether it is related or not related

 

 23   to their own research.  If it is, then it takes

 

 24   them back to make some comments to the original

 

 25   report that I submitted on my study.  So, there is

 

                                                               115

 

  1   a page that allows the other PIs to come back in

 

  2   and give additional information.

 

  3             This doesn't project very well and I

 

  4   apologize, but all this data then can be captured

 

  5   in different ways.  In this particular page there

 

  6   is data on one study and all the adverse events

 

  7   that have been reported on that study within, I

 

  8   think, a six-month period.  Each one of those cells

 

  9   can be manipulated to provide you different ways of

 

 10   looking at the data.  So, you could ask the data to

 

 11   be cut only at grade 3 or grade 4 or only deaths on

 

 12   that particular study.  You can ask the system to

 

 13   report all deaths on all patients across three

 

 14   studies to see if there are complementary problems,

 

 15   and things like that.

 

 16             So, this is where we are right now.  We

 

 17   established this about 18 months ago.  The next

 

 18   phase of this project is actually now beginning to

 

 19   mine the data so that we can create some normative

 

 20   rules of when we should be setting lines that raise

 

 21   red flags that we should pay more attention to.

 

 22   So, I think that is the strength of this, that now

 

 23   it unifies it in a certain way so that now we can

 

 24   go back and make some sense of all the data, and I

 

 25   think with that I will stop.  Thank you.

 

                                                               116

 

  1             Oh, obviously I didn't thank everybody

 

  2   that was part of the team.  Don Workman, our IRB

 

  3   administrator, was very involved with this.  Donna

 

  4   Hogan, from the IRB office, is in charge of the AE

 

  5   reporter.  Then, two individuals from clinical

 

  6   informatics were the ones who put all these ideas

 

  7   to work.  Thanks.

 

  8             Now I think we have some time for

 

  9   questions before we go into the discussion.

 

 10                       Committee Discussion

 

 11             DR. HIRSCHFELD:  I have a question for Dr.

 

 12   Anderson.  Dr. Santana discussed the goal of the

 

 13   project at St. Jude to get some normative data on

 

 14   what types of events one can expect and, perhaps by

 

 15   implication, what needs to be monitored and what

 

 16   doesn't need to be monitored, and when things do

 

 17   occur how serious they are.  Does the NCI have such

 

 18   a program?  If it does, are there any analyses that

 

 19   you are able to share?  Or Dr. Smith could answer

 

 20   the question.

 

 21             DR. ANDERSON:  I am not aware of a

 

 22   specific program for pediatric oncology, you know,

 

 23   with the AdEERS system for bringing in information.

 

 24   There is data trial by trial, especially for IND

 

 25   agents.  That sort of information is accumulated. 

 

                                                               117

 

  1   But to provide sort of a baseline level of here is

 

  2   what to look for over time, I don't know that that

 

  3   is a specific project that is under way right now.

 

  4             DR. SANTANA:  Yes, in fairness to the

 

  5   question, we are not doing that right now.  We have

 

  6   the capability based on this project after we have

 

  7   been into it 18 months because we thought about

 

  8   that when we tried to build the electronic format.

 

  9   We now have the ability to do that but, in fairness

 

 10   to the question, we have not done that.  We are

 

 11   just establishing the data and, hopefully, at some

 

 12   point we will begin to analyze it once we have

 

 13   enough data to make some sense out of it.  We are

 

 14   really only, particularly right now, focusing on

 

 15   the St. Jude studies, studies where we are the

 

 16   primary sponsor.

 

 17             DR. SMITH:  Steve, as Barry said, we do

 

 18   have the AdEERS system that is an electronic

 

 19   reporting system.  So, you know, there is the

 

 20   capability if there is a question about cardiac

 

 21   toxicity or other organ toxicity to pull up all of

 

 22   those reports for a particular toxicity.  But in

 

 23   terms of what to look for, you know, if the

 

 24   question is what toxicities are occurring in what

 

 25   types of trials, then the Phase I and Phase II

 

                                                               118

 

  1   databases of the Phase I Consortium and the

 

  2   Pediatric Brain Tumor Consortium are more relevant

 

  3   because if the AE reporting is being done

 

  4   correctly, then it is the unexpected events.  You

 

  5   know, what you would really be interested in is the

 

  6   whole universe of events and, as well, the

 

  7   denominator of how many patients were in those

 

  8   trials.  So, I think I would approach one of the

 

  9   consortia for early phase trials or COG for later

 

 10   phase trials if the question was what type of

 

 11   events are occurring, how frequently they are

 

 12   occurring, etc.

 

 13             DR. SANTANA:  Dr. Przepiorka?

 

 14             DR. PRZEPIORKA:  Back to Steve, if I could

 

 15   turn the question right back to you, does the FDA

 

 16   have enough information or a database on SAEs in

 

 17   pediatric trials to actually do that same study?

 

 18             DR. HIRSCHFELD:  Short answer?  No.  We

 

 19   would like to but we don't have a database that

 

 20   captures premarketing adverse events.  We only have

 

 21   a database for postmarketing adverse events.  That

 

 22   is mined in a fairly rigorous and maybe even

 

 23   imaginative way to look at frequencies of what one

 

 24   can expect but, again, it hasn't been examined

 

 25   sufficiently on the basis of pediatrics and, even

 

                                                               119

 

  1   more specifically, on pediatric oncology.  So, we

 

  2   don't have the data and that is one of the issues

 

  3   and one of the reasons for having this discussion

 

  4   this morning.

 

  5             DR. DAGHER:  Dr. Santana, another question

 

  6   about your presentation which also may impact on

 

  7   the NCI perspective, one of the challenges you

 

  8   identified was the situation where a patient is

 

  9   enrolled on several studies at the same time.  I am

 

 10   curious to know how often that happens and whether

 

 11   that is somewhat unique to St. Jude, or is that

 

 12   something that you also see across pediatric

 

 13   studies that NCI supports?

 

 14             DR. SANTANA:  The way the system is

 

 15   designed is that it will pick any protocol that the

 

 16   patient is still currently enrolled on.  It doesn't

 

 17   mean the patient is on active therapy on those

 

 18   other studies; it may be that they are in follow-up

 

 19   for those other studies for example but the patient

 

 20   has not been taken off those additional studies.

 

 21   We did that on purpose in terms of thinking outside

 

 22   the box, that if there were long-term issues with

 

 23   patients that had been enrolled on other studies

 

 24   and then you began to see trends that were

 

 25   complementary to a group of studies that together

 

                                                               120

 

  1   created something in the future, we could go back

 

  2   and capture that.

 

  3             So, your point is well taken.  The primary

 

  4   study that is generating the adverse events is many

 

  5   times the active study that the patient is being

 

  6   treated on.  But we also wanted to make sure that

 

  7   we were able to capture data on studies where the

 

  8   patient was not actively receiving therapy but was

 

  9   still technically enrolled on that study.

 

 10             Having said that, we also wanted to

 

 11   capture non-therapeutic trials so it will list any

 

 12   trial.  It won't make any distinction whether it is

 

 13   therapeutic or non-therapeutic up front.

 

 14             DR. DAGHER:  Supportive care--

 

 15             DR. SANTANA:  Yes.  On the last page there

 

 16   was one trial which was a behavioral medicine trial

 

 17   on which the patient had been enrolled that had

 

 18   nothing to do with the primary therapeutic trial,

 

 19   and that showed up too.

 

 20             DR. HIRSCHFELD:  May I ask for just one

 

 21   more clarification on your presentation, Dr.

 

 22   Santana?  You said that the system in use at St.

 

 23   Jude will bring up the relevant definitions for a

 

 24   serious adverse event, unexpected, etc.  What is

 

 25   the source of those definitions?  There are several

 

                                                               121

 

  1   places that are source documents, including ICH

 

  2   documents.

 

  3             DR. SANTANA:  I think what we did was an

 

  4   amalgam of the different definitions and tried to

 

  5   make it into a definition that people could

 

  6   understand without having to pick up a dictionary

 

  7   or call the IRB administrator.  So, it was really

 

  8   looking at all those documents and coming up with

 

  9   some definitions that were kind of a semi-practical

 

 10   way that people could relate to and then choose the

 

 11   right box.  Dr. Grillo-Lopez?

 

 12             DR. GRILLO-LOPEZ:  I have a suggestion for

 

 13   future meetings on this subject, and that is to

 

 14   invite a representative from the pharmaceutical

 

 15   industry to make a presentation because although

 

 16   you might argue that the FDA knows very well how

 

 17   the pharmaceutical industry functions in terms of

 

 18   monitoring adverse event reporting, on the other

 

 19   hand, others around this table and others

 

 20   participating in the Webcast or viewing the tapes

 

 21   later on might not.  The fact is that there is

 

 22   extensive experience with clinical trial monitoring

 

 23   in the pharmaceutical industry and, likewise, with

 

 24   adverse event reporting.  Although I see a great

 

 25   parallel and even consistency in terms of the

 

                                                               122

 

  1   procedures and methods that are used in your

 

  2   institution representing an academic experience,

 

  3   and at the NCI particularly with the cooperative

 

  4   groups, there are some points that are different

 

  5   and that would merit discussing and presenting

 

  6   because they might present opportunities for

 

  7   improvement.

 

  8             DR. SANTANA:  Are you in a position to

 

  9   highlight some of those points?

 

 10             DR. GRILLO-LOPEZ:  Well, one thing that

 

 11   just came up in the discussion was the subject of

 

 12   denominators.  Certainly, when you are conducting

 

 13   research with a new therapeutic agent the database

 

 14   at that pharmaceutical company contains the most

 

 15   information regarding the safety experience with

 

 16   that agent at any given point in time.  Of course,

 

 17   all of that database is transferred to the FDA as

 

 18   required.  But investigators participating in

 

 19   multicenter trials could certainly call the project

 

 20   clinician who would have access, through his

 

 21   biometrics group, to that database and would be

 

 22   able to provide information about what the

 

 23   experience has been with other events of that

 

 24   nature.

 

 25             DR. HIRSCHFELD:  Dr. Santana, if I may

 

                                                               123

 

  1   just respond to the initial suggestion, and I want

 

  2   to thank Dr. Grillo-Lopez, one of the reasons you

 

  3   are at the table is to provide that.  Previously at

 

  4   the meetings of this committee we had multiple

 

  5   representatives from the pharmaceutical industry

 

  6   and had routinely asked for presentations but there

 

  7   was a policy decision made outside the group that

 

  8   you see here today to restrict that.  So, since you

 

  9   are new to the process--had you been involved

 

 10   earlier you would have seen what you are

 

 11   suggesting--maybe you can help us restore that

 

 12   previous mode of interaction because we found it

 

 13   helpful also.

 

 14             DR. GRILLO-LOPEZ:  Yes, that was an

 

 15   unfortunate decision and I, of course, didn't know

 

 16   about that.  I think it is a three-legged stool or

 

 17   a triangle, as you were saying, with the

 

 18   participation, on the one hand, of the NCI and

 

 19   cooperative groups particularly, individual

 

 20   academic institutions and the pharmaceutical

 

 21   industry as sponsors in conducting research.  We

 

 22   should not forget that third leg of the stool

 

 23   because a lot of the research that is conducted

 

 24   with new agents particularly is sponsored by the

 

 25   pharmaceutical industry and the pharmaceutical

 

                                                               124

 

  1   industry holds the databases for the results of

 

  2   that research, and one particular institution may

 

  3   have had a lot of experience with a new agent but

 

  4   not necessarily all of the experience because many

 

  5   other institutions might be participating and they

 

  6   may not be communicating between themselves but

 

  7   certainly the database of the pharmaceutical

 

  8   company holds all of that information.

 

  9             DR. SANTANA:  Dr. Adamson?

 

 10             DR. ADAMSON:  A couple of comments, first,

 

 11   I want people to be aware that what Dr. Santana

 

 12   presented, which I think is something academic

 

 13   institutions should strive for, is not the norm.

 

 14   Most institutions are many steps behind what St.

 

 15   Jude has done and is capable of doing, and in most

 

 16   institutions what you are looking at are piles and

 

 17   piles of paper.  So, your colleagues are to be

 

 18   commended on beginning to address what is a problem

 

 19   for all academic institutions.

 

 20             I wanted to comment that I think the

 

 21   current SAE and AE mechanism--and this will echo

 

 22   and build upon what Victor said--has some

 

 23   significant flaws.  I mean, we can be inundated

 

 24   with reports that we cannot interpret, and what I

 

 25   would say is that the large majority of external

 

                                                               125

 

  1   reports, when it comes to the cover letter,

 

  2   "because of regulation blah, blah, blah, you are

 

  3   required to submit this to your IRB"--the large

 

  4   majority of those reports, as a member of an IRB as

 

  5   well as an investigator, one cannot interpret.  It

 

  6   gets down to knowing the denominator and you said

 

  7   there are large databases but the problem is you

 

  8   need real-time access to that database in order to

 

  9   interpret it.  There is too large a line of these

 

 10   reports coming in for the investigator to call and

 

 11   track down every report--is this relevant?  Has the

 

 12   risk/benefit ratio really changed for my patient?

 

 13             Rick said earlier we should try to focus

 

 14   on pediatrics so I will.  As one moves

 

 15   forward--because this is the problem and it is not

 

 16   limited to pediatrics but is a problem across the

 

 17   board that one can't interpret the large majority

 

 18   of these reports and we are fooling ourselves if we

 

 19   think simply by submitting the document to the IRB

 

 20   you have fulfilled your obligation.  That is not

 

 21   improving patient safety.  You may have fulfilled

 

 22   the regulatory obligation but you have done nothing

 

 23   to improve patient safety.  We need access to the

 

 24   type of data you referred to that industry has to

 

 25   interpret this.

 

                                                               126

 

  1             To focus on pediatrics, I will give you an

 

  2   example.  We did an industry-sponsored study and

 

  3   there were many studies of this investigational

 

  4   agent.  The large majority of reports were about

 

  5   myocardial infarction in a 76 year-old.  That is

 

  6   important but it is not particularly relevant to

 

  7   the pediatric population.  So, when we move forward

 

  8   and ask for data, I think we need to have some

 

  9   depth to that data, that is, not only the frequency

 

 10   of the event but somehow to categorize what

 

 11   population that event is occurring in.  Because if

 

 12   an event is occurring in a 30 year-old--and my mark

 

 13   of what I think is young is continually shifting

 

 14   upwards--

 

 15             [Laughter]

 

 16             --but if an event were to occur in a 30

 

 17   year-old you might spend a little more time looking

 

 18   at that event as far as, you know, was it a

 

 19   cardiovascular event relative to someone who is

 

 20   more elderly.  So, I would hope that one looks at

 

 21   the regulations and makes it that you don't just

 

 22   send the report, but the report has to be in

 

 23   context and the context is what is happening

 

 24   globally with the safety of this drug, focusing on

 

 25   its particular toxicity, but within that have the

 

                                                               127

 

  1   depth to say this is the breakdown of the

 

  2   population that we are looking at.  I mean, you

 

  3   don't need to get it down to all 12 year-olds, all

 

  4   13 year-olds or all 20 year-olds but give us some

 

  5   sense of what is happening.  Otherwise, I don't

 

  6   think we are doing anything for patient safety in a

 

  7   meaningful way for the large majority of these

 

  8   reports.

 

  9             DR. KODISH:  This is Eric, in Cleveland.

 

 10   I hope my timing is okay and you can hear me.

 

 11             DR. SANTANA:  Yes, Eric, go ahead.

 

 12             DR. KODISH:  Thanks.  I want to add an

 

 13   idea to Peter's idea which I think is very

 

 14   important and relates to the point I was trying to

 

 15   make about regulation actually harming patient

 

 16   safety on some level.

 

 17             A 76 year-old who has a myocardial

 

 18   infarction on a drug that we are testing in a

 

 19   pediatric population compared to a 30 year-old

 

 20   compared to a 20 year-old I think gives us the

 

 21   ability to maybe, rather than contextualize which

 

 22   would be great--ut maybe a more simple idea is to

 

 23   provide some sort of sorting function so that we

 

 24   are not just, for regulatory or prevention of

 

 25   litigation, trying to download all of these reports

 

                                                               128

 

  1   to our IRBs to say that we have fulfilled

 

  2   regulatory requirements, but that at some level

 

  3   there could be a sorting function so that the

 

  4   events that are going to be relevant to children

 

  5   are presorted, if you will, and not disseminated

 

  6   across the country automatically. I think we do

 

  7   need to be concerned about the paradoxical effect

 

  8   of everyone feeling that because we have filed all

 

  9   these adverse event reports that everything is

 

 10   going to be okay.

 

 11             DR. SANTANA:  Eric, just to play devil's

 

 12   advocate with your comment and Peter's comment, who

 

 13   defines what is relevant to our population?  It is

 

 14   us.  And, I think that is probably why we are here

 

 15   today.  We have to define in our studies, either

 

 16   prospectively when the study is being created based

 

 17   on what we know about the agent or whatever is

 

 18   going to happen in the study or during the conduct

 

 19   of the study as we review things--we are the ones

 

 20   that have to define what triggers that it is a

 

 21   pediatric issue that we need to address.  If not,

 

 22   then we just rely on these big data warehouses that

 

 23   have data that are not relevant, but we have to

 

 24   define the relevance up front or during the conduct

 

 25   of the study.

 

                                                               129

 

  1             DR. KODISH:  I agree with that and I think

 

  2   that maybe the discussion could focus on how we

 

  3   sort those that are and those that aren't, maybe

 

  4   starting with something as simple as an age

 

  5   cut-off.

 

  6             DR. SANTANA:  Well, one of the things that

 

  7   Barry mentioned in his presentation, and in

 

  8   retrospect I wish he had given more discussion to

 

  9   his point, was this issue of how some of the

 

 10   consortia--and it is the PBTC Phase I group or

 

 11   maybe it is the COG--that those committees

 

 12   electronically and telephonically and through

 

 13   computers meet on a regular basis and they review

 

 14   real-time data of those patients that are on Phase

 

 15   I studies.  I presume, and I think correctly so,

 

 16   that there also is, as part of that review, the

 

 17   toxicity and the adverse events occurring in those

 

 18   patients.  So, that whole arm of this process,

 

 19   which we didn't discuss in great detail, I think is

 

 20   very strong because it relies in part on the

 

 21   research team to very actively monitor this in

 

 22   their own hands.

 

 23             We have to have checks and balances

 

 24   through other groups too but the beauty of that is

 

 25   that it allows the research team who is actually

 

                                                               130

 

  1   conducting the research in real time to be able to

 

  2   communicate and evaluate these and then

 

  3   prospectively, even as the study is being

 

  4   conducted, define what are the parameters that

 

  5   trigger the normative data that we are looking for

 

  6   because in reality it is an experiment.  Until we

 

  7   do it we are really not going to know the whole

 

  8   scope of things that may happen.  We kind of can

 

  9   predict based on what are the things that may

 

 10   happen or what are the things that would really

 

 11   worry us.  Right?  If somebody dies we all worry,

 

 12   or if something unexpected occurs we all worry.

 

 13   But for the majority of things, things are

 

 14   happening and it creates a lot of noise.  I agree

 

 15   with you, Peter, it creates a lot of noise.  So, I

 

 16   think we have to go back to the research team and

 

 17   address what their role and responsibility is to

 

 18   help us at the other end figure out how this data

 

 19   may be interpreted or may be incorporated.

 

 20             DR. ADAMSON:  I think you are right.

 

 21   Phase I, in many respects, is somewhat easier

 

 22   because the data is being monitored in real time

 

 23   and the numbers are small.  I think it is a

 

 24   multi-level process.  It begins with the treating

 

 25   institution and the team at that institution

 

                                                               131

 

  1   recognizing and identifying the event and reporting

 

  2   it to the study principal investigator.

 

  3             What we do in our consortium is once it is

 

  4   to the study investigator it immediately comes to

 

  5   us and then on a weekly basis all the events on

 

  6   every study are reviewed.  What we have the ability

 

  7   to do and what we focus on is that we don't just

 

  8   look at the serious ones because the serious ones

 

  9   are usually pretty straightforward.  We look at the

 

 10   non-serious toxicities to look for trends because

 

 11   we are doing a dose escalation and so we want to

 

 12   know.  Okay, we are starting to see some grade 2s

 

 13   in an area that wasn't described that are not

 

 14   triggering any alarms but, in fact, maybe we need

 

 15   to do more careful monitoring of hepatic function

 

 16   because we are seeing a lower level of toxicity.

 

 17   So, it is a multi-level review but we have the

 

 18   ability to look at all the toxicities on a study as

 

 19   a function of dose, as a function of severity and

 

 20   that gives us the context to interpret it.

 

 21             DR. SANTANA:  Dr. Grillo and then Dr.

 

 22   Carome.

 

 23             DR. GRILLO-LOPEZ:  I find that we are

 

 24   talking about adverse events in general but also

 

 25   about serious adverse events and perhaps not always

 

                                                               132

 

  1   making a distinction about the different reporting

 

  2   requirements for those.  Certainly, in the

 

  3   pharmaceutical industry we collect each and every

 

  4   adverse event but the reporting requirements are

 

  5   different if it is a serious adverse event.  It

 

  6   might help if someone from the FDA would just

 

  7   summarize what the requirements are for reporting

 

  8   to an IRB and reporting to the FDA.

 

  9             DR. HIRSCHFELD:  The definitions are

 

 10   essentially ICH definitions, International

 

 11   Conference on Harmonization, that the FDA adopts.

 

 12   The requirement essentially is if it is serious an

 

 13   unexpected according to both triggers, then there

 

 14   has to be what is called a rapid report filed.

 

 15   That can be filed by a number of mechanisms.  The

 

 16   time frame typically is within 15 days and

 

 17   sometimes, depending on the circumstance, can be 7

 

 18   days.  But that is still not what could be called

 

 19   real time.  It is essentially informing.  All other

 

 20   adverse events do not have to be reported to the

 

 21   FDA, other than in the annual reports which are

 

 22   required.  The annual reports are due within 90

 

 23   days of the initial filing of the IND.

 

 24             DR. GRILLO-LOPEZ:  How about to IRBs?

 

 25             DR. HIRSCHFELD:  The IRB requirements work

 

                                                               133

 

  1   on multiple levels.  So, the IRB can set their own

 

  2   policy but in the FDA regulations, in 21 CFR 50,

 

  3   the reporting requirements for IRBs parallel those

 

  4   of reporting to the FDA.

 

  5             DR. GRILLO-LOPEZ:  If I may, in that vein

 

  6   I would make the point that we have to be very

 

  7   precise, very specific and very timely in reporting

 

  8   serious an unexpected adverse events.  At the other

 

  9   extreme, there is a multitude of minor events that

 

 10   are still adverse events and need to be in the

 

 11   database at some point without creating this

 

 12   backlog, this bureaucratic mass of paper and

 

 13   electronic data coming at you without denominators,

 

 14   which doesn't make much sense at any one given

 

 15   point in time for one patient.

 

 16             However, many times we find at the end of

 

 17   the development of a new therapeutic that when we

 

 18   have to put together the documentation to submit to

 

 19   the FDA, one of the things that we, in industry,

 

 20   have to do is to do an analysis across all of the

 

 21   experience with that agent, Phase I, II, III, all

 

 22   of the studies ever done, which is called the

 

 23   integrated summary of safety.  Many times it is

 

 24   only then that certain trends become significant

 

 25   that were not significant earlier on when you only

 

                                                               134

 

  1   had the Phase I or the Phase II experience.  That

 

  2   is why it is important to report each and every

 

  3   adverse event but not necessarily make it a

 

  4   bureaucratic jungle where you just get so entangled

 

  5   in paper and data that it doesn't make sense at any

 

  6   one given point in time.

 

  7             DR. SANTANA:  Dr. Grillo, you represent

 

  8   the pharmaceutical aspects of this.  As somebody

 

  9   from that group specifically focusing on pediatric

 

 10   oncology issues, how would you advise your group of

 

 11   things that we need to have access to, and in what

 

 12   time lines would you advise your group that we need

 

 13   to have access to those data so that we can

 

 14   complement that with what we want to do?

 

 15             DR. GRILLO-LOPEZ:  I may not be the right

 

 16   person to respond to that question because I am an

 

 17   adult oncologist, not pediatric oncologist.  In

 

 18   fact, in over 20 years in industry, I never did a

 

 19   pediatric study, ever.  So, I have zero experience

 

 20   and I have to be the first one to admit to that,

 

 21   other than my rotation through pediatric oncology

 

 22   when I was a fellow.

 

 23             But I think there is a variety of ways and

 

 24   systems and procedures that the pharmaceutical

 

 25   industry utilizes to follow-up, collect and be able

 

                                                               135

 

  1   to analyze adverse events.  It begins with the case

 

  2   report forms coming in from the different sites

 

  3   participating in a multicenter study.  I can tell

 

  4   you that for all of the studies that I was ever

 

  5   related with, I would personally look at each and

 

  6   every piece of paper coming in from the different

 

  7   sites, or the safety officer responsible within my

 

  8   group would do that even before it went into the

 

  9   database.  So, if there was a major red flag that

 

 10   was apparent even just from the experience in one

 

 11   patient, we would see that.  Of course, immediately

 

 12   that was entered into the database and periodically

 

 13   we would print out tabulations that would indicate

 

 14   if there was any trend that was becoming obvious.

 

 15             So, there is a variety of checks and

 

 16   balances that are in place within the

 

 17   pharmaceutical industry to follow-up on these

 

 18   issues.  Again, I would suggest that investigators

 

 19   who are participating in multicenter pharmaceutical

 

 20   industry sponsored studies, that their point of

 

 21   access or one point of access might be the project

 

 22   clinician within the pharmaceutical company who is

 

 23   the person responsible and/or the safety officer

 

 24   within that company when issues arise or questions

 

 25   arise regarding a specific adverse event.

 

                                                               136

 

  1             DR. SANTANA:  Dr. Carome?

 

  2             DR. CAROME:  I will just note a couple of

 

  3   things.  I think it is important for this

 

  4   subcommittee to be aware that this discussion is

 

  5   occurring elsewhere.  The Secretary's Advisory

 

  6   Committee on Human Research Protections had a panel

 

  7   on adverse event reporting and they are going to

 

  8   continue that discussion at their next meeting.

 

  9   They had a panel in December and they are going to

 

 10   continue the discussion in their March meeting,

 

 11   coming up in a couple of weeks.  And the discussion

 

 12   is exactly the same.  I mean, the types of comments

 

 13   being articulated are verbatim what you hear

 

 14   repeatedly.

 

 15             I think the Department recognizes that

 

 16   there is a need to make adverse event reporting

 

 17   more meaningful and less burdensome in order to

 

 18   better protect human subjects, and there are

 

 19   ongoing discussions between our office, the FDA,

 

 20   NIH and other federal departments and agencies on

 

 21   how best to do that.  So, it is recognized to be a

 

 22   problem and developing strategies is complex but we

 

 23   believe important.

 

 24             If you look at our regulations, just the

 

 25   HHS regulations CFR 46, there is no adverse event

 

                                                               137

 

  1   reporting requirement.  There is a requirement for

 

  2   reporting what are called unanticipated problems

 

  3   involving risk to others.  It is our view that most

 

  4   adverse events that occur in clinical trials do not

 

  5   fall into that category and, therefore, under our

 

  6   regulations the vast majority of adverse event

 

  7   reports do not need to be reported under our

 

  8   regulations.  Those that we particularly care

 

  9   about, and we have articulated this at the

 

 10   Secretary's advisory committee in December, are

 

 11   those that represent unexpected, serious harms to

 

 12   subjects, which are words that come from another

 

 13   part of our regulation.  Those are the types of

 

 14   events we think should get to IRBs and that we care

 

 15   most about.

 

 16             DR. SANTANA:  So, Mike, where do you think

 

 17   the confusion comes that all these reports are

 

 18   being generated and submitted to IRBs?  Where do

 

 19   you think the communication breakdown is in terms

 

 20   of what the regulatory agencies want versus what

 

 21   the sponsors or we, as investigators, see that you

 

 22   guys want and need to comply with?

 

 23             DR. CAROME:  There are probably multiple

 

 24   reasons.  It is clear to us, and I think to others,

 

 25   that the greatest burden comes from these external

 

                                                               138

 

  1   adverse events that don't occur at your site but,

 

  2   because we do research at multiple sites, the

 

  3   sponsors deliver those reports or ask that they be

 

  4   delivered to the investigators at all sites.  So,

 

  5   now we have 100 IRBs maybe receiving the same event

 

  6   so it is those external events that are being

 

  7   multiplied to multiple IRBs where the burden has

 

  8   been articulated to us as being most severe, and if

 

  9   the letter reads that under the regulations you

 

 10   must deliver these to your IRB, that is certainly

 

 11   one source.  It is not our regulations that are

 

 12   demanding that and I would posit that a close look

 

 13   at the FDA regulations probably doesn't justify all

 

 14   those events going to the IRB as well.  But FDA

 

 15   would have to comment on that.  So, that is one.

 

 16             I think it is driven by fears of

 

 17   litigation liability.  You know, who makes this

 

 18   initial assessment about unexpected and serious?

 

 19   We think that at one level the sponsor and the

 

 20   investigator can be doing that.  There are some who

 

 21   think those are conflicted parties and maybe we

 

 22   need an independent body making those decisions so

 

 23   people are driven to having an independent body be

 

 24   the IRB looking at them.  So, I think there are

 

 25   multiple reasons.  Those are a couple that I would

 

                                                               139

 

  1   highlight as perhaps driving it.

 

  2             DR. SANTANA:  Rick, do you want to comment

 

  3   on the FDA?

 

  4             DR. PAZDUR:  Well, I just want to comment

 

  5   in general.  Could there be attempts to try to give

 

  6   investigators more guidance on specifically what

 

  7   needs to be reported?  I think there is a tendency

 

  8   to report a lot to cover oneself because we don't

 

  9   have good guidance on exactly what those words

 

 10   mean.  Maybe we need to look into that.  You know,

 

 11   if you go over your phrase that you gave, there is

 

 12   a lot of interpretation here and somebody could say

 

 13   that it might be the index case; they are not even

 

 14   sure of the attribution issue, and I wonder if we

 

 15   really need to give more guidance to perhaps cut

 

 16   down on some of this.  I don't know, do you want to

 

 17   comment on that?

 

 18             DR. CAROME:  I think for us, we believe

 

 19   guidance is essential and it is the most important

 

 20   step.  We have had discussions with FDA.  We are

 

 21   prepared to draft guidance that articulates in more

 

 22   detail what I just articulated to you and I

 

 23   previously articulated at the Secretary's Advisory

 

 24   Committee on Human Research Protections in

 

 25   December.  But we think, yes, guidance is the

 

                                                               140

 

  1   important step.  We think because adverse events

 

  2   are primarily referenced in FDA regulations the

 

  3   guidance needs to come out of both of our offices

 

  4   or entities.

 

  5             DR. SANTANA:  Dr. Smith, I think you had

 

  6   your hand up?

 

  7             DR. SMITH:  One point, and, Victor, I

 

  8   think you made it, there is over-reporting of

 

  9   adverse events, expedited adverse events, despite

 

 10   FDA's stated requirements, despite their statements

 

 11   in protocols of what does require expedited

 

 12   reporting.  So, I think one of the initiatives that

 

 13   we want to undertake in the next few months is an

 

 14   educational initiative to try to limit the

 

 15   over-reporting of things that, in fact, just do not

 

 16   require regulatory reporting.  So, this will

 

 17   decrease some of the burden at the institutional

 

 18   level.

 

 19             It doesn't address the issue, however,

 

 20   that Peter raised about when you get a letter from

 

 21   a company saying that this event occurred.  I

 

 22   wonder if there is a role for the Phase I

 

 23   Consortium itself or the COG itself to play the

 

 24   filter role that Eric Kodish was talking about in

 

 25   terms of saying we have reviewed this, and our

 

                                                               141

 

  1   recommendation to IRBs, when they look at it, is to

 

  2   say this isn't applicable for pediatrics.

 

  3             DR. ADAMSON:  We are actually now doing

 

  4   that, Malcolm, when it comes, you know, a COG

 

  5   trial.  When we disseminate it we usually give a

 

  6   recommendation that, in our view, this does not

 

  7   change risk/benefit or, in our view, it does change

 

  8   the risk/benefit ratio and it should be reported.

 

  9   So, we try to put it into context but, of course,

 

 10   every investigator has the ability to interpret the

 

 11   data and make their own decisions.

 

 12             DR. SANTANA:  Dr. Przepiorka, you had your

 

 13   hand up?

 

 14             DR. PRZEPIORKA:  Yes, I clearly remember

 

 15   sitting through multiple discussions at the

 

 16   initiation site visits with sponsors regarding the

 

 17   definition of an SAE, and I recall a few years ago,

 

 18   after the incident at Penn and the FDA sent that

 

 19   Webcast to all the academic institutions with a

 

 20   long, drawn-out discussion on what is an SAE, and I

 

 21   sat here and I think we listed them, although I

 

 22   didn't see them on any of the slides this morning.

 

 23   I won't go through them but I don't see any that is

 

 24   very specific to pediatrics and I am wondering if

 

 25   there is any SAE that should be added to the list

 

                                                               142

 

  1   specifically for pediatric groups.  I am thinking

 

  2   about long-term cognitive dysfunction or something

 

  3   like that.

 

  4             DR. SANTANA:  Ruth?

 

  5             MS. HOFFMAN:  I was just going to mention

 

  6   that I sit on the IRB at Children's National

 

  7   Medical Center, as well as the DSMB board there,

 

  8   and from a lay perspective it is very difficult to

 

  9   get lay people to continue with the responsibility

 

 10   because of the burden of time commitment.  There is

 

 11   no monetary compensation.  I don't get paid and I

 

 12   am not an employee of Children's National Medical

 

 13   Center.  I spend three days a month totally related

 

 14   to IRB-related work between the protocols and the

 

 15   SAEs and AEs.  I mean, it is just a stack of paper

 

 16   and usually the check-off is that the AE has

 

 17   nothing to do with the protocol at all and, you

 

 18   know, maybe you can eliminate that whole column

 

 19   and, again, reduce the workload.  But, I mean, they

 

 20   have a very hard time to even recruit members from

 

 21   the community and that is a requirement of the HHS,

 

 22   to have a lay person on the committee.  So, the

 

 23   guidance document would be great.  It would

 

 24   certainly help from our perspective as well.

 

 25             DR. SANTANA:  Dr. Grillo-Lopez?

 

                                                               143

 

  1             DR. GRILLO-LOPEZ:  I would suggest that

 

  2   further guidance is not necessary, that what we

 

  3   need is education.  The guidance that is already

 

  4   provided by the FDA is very specific and very clear

 

  5   as to what is a serious and unexpected adverse

 

  6   event, and what we need is for those involved in

 

  7   research, and particularly at the IRB level, to

 

  8   have an understanding of what that means.  I think

 

  9   it is education.  Generating one more document to

 

 10   file away does not help anyone.

 

 11             DR. SANTANA:  Peter?

 

 12             DR. ADAMSON:  I agree that the FDA

 

 13   guidance on the definition of an SAE is clear.

 

 14   What I think the point is, is that it is not

 

 15   particularly functional in that it is generating an

 

 16   incredible amount of paperwork for institutions.

 

 17   What we get are, indeed, SAEs by the definition.

 

 18   That information I don't think is improving patient

 

 19   safety and that is why I would actually agree that

 

 20   we need to re-look at what we are requiring to be

 

 21   reported to IRBs across the country because it is

 

 22   not only a multi-institutional trial, it is when

 

 23   you have multiple trials of an investigational drug

 

 24   that affects all those trials.

 

 25             DR. SANTANA:  Dr. Keegan?

 

                                                               144

 

  1             DR. KEEGAN:  Yes, I was wondering if we

 

  2   could go back to the concept that was discussed

 

  3   before about having a central body that looks at

 

  4   all the adverse events because, as you say, every

 

  5   individual institution is going to be unable to

 

  6   look at a single adverse event out of context with

 

  7   the rest of the data.  So, to what extent are there

 

  8   really plans in place to have a central point that

 

  9   has all the data that could make reasonable

 

 10   interpretations that have people with the

 

 11   background information who could interpret the

 

 12   adverse event information in the context of animal

 

 13   and nonclinical studies and other things to make

 

 14   relevant decisions?  Because you mentioned that in

 

 15   the instance of the consortium but it doesn't seem

 

 16   that that is a general theme.  To some extent, I

 

 17   don't think any one individual is ever going to be

 

 18   able to make a conclusion on the index case but we

 

 19   certainly can't ignore the index cases because that

 

 20   also puts patients at risk.

 

 21             DR. SANTANA:  Patricia, in follow-up to

 

 22   that comment, what kind of body were you thinking

 

 23   of?  What ideal body, if you had to come up with

 

 24   that, would you propose?

 

 25             DR. KEEGAN:  Well, it sounds like that is

 

                                                               145

 

  1   sort of the model that the consortia are working on

 

  2   and I thought maybe there could be more discussion

 

  3   of whether it could be that sort of model, where

 

  4   the consortium looks at adverse events and then

 

  5   sends out their interpretation as a central

 

  6   repository analysis, much like a medical monitor

 

  7   would do at a drug company to perform that same

 

  8   function.

 

  9             DR. ADAMSON:  Well, I think it is easier

 

 10   for smaller studies, and the key thing is you have

 

 11   access to all the data.  So, when we do an

 

 12   NCI-sponsored study where the NCI is cross-filed

 

 13   there is a drug monitor at the NCI that has access

 

 14   to all the data and, in fact--correct me if I am

 

 15   wrong, Barry--usually when an AE comes out there

 

 16   also is a recommendation of an interpretation when

 

 17   it happens.  I can't say that is the case uniformly

 

 18   for industry-sponsored trials.  But the multiplying

 

 19   effect I think is a difficult effect for when

 

 20   events are occurring really that are distantly

 

 21   related to the study that you are doing.

 

 22             DR. SANTANA:  I think the advantage that

 

 23   we have in pediatric oncology is that it is a

 

 24   smaller universe and most pediatric, if not all

 

 25   pediatric oncology studies are really conducted in

 

                                                               146

 

  1   the list that Barry showed, plus a few others.

 

  2   Right?  So, we are a much smaller universe so that

 

  3   if we adopted a model similar to what is happening

 

  4   in the Phase I consortium and expanded that to all

 

  5   the participants in those groups because it is a

 

  6   small universe, we could at least set that model

 

  7   and see if it works for us.  Because that is what

 

  8   we are really here for, right?  For pediatric

 

  9   oncology, not to ignore or belittle the other

 

 10   important issues that are occurring with adults but

 

 11   we have that advantage and maybe we should think of

 

 12   that model as a test case for reviewing adverse

 

 13   event reports to make it more functional and

 

 14   timely.

 

 15             To me, the issue is time.  So, what if

 

 16   something happened six months ago?  It doesn't help

 

 17   my patient who is on the study now.  Right?  So,

 

 18   maybe we have that advantage.  We are a small

 

 19   group.  Malcolm?

 

 20             DR. SMITH:  The other possibility is, Pat,

 

 21   we are at the earliest stages of setting up a

 

 22   pediatric central IRB and so, you know, could that

 

 23   be a body that is somehow constituted so that it

 

 24   could play that role nationwide and then other IRBs

 

 25   could use that information if they chose to?

 

                                                               147

 

  1             DR. SANTANA:  Since you raised the issue I

 

  2   am going to try to explore it a little bit further.

 

  3   There has been some recent discussion I think in

 

  4   some of the things I have been reading about

 

  5   whether DSMB should play some of this role.  Do you

 

  6   want to comment on that?

 

  7             DR. PAZDUR:  I would just say that I had a

 

  8   side conversation with Susan, here, and one of the

 

  9   issues is that usually DSMBs are single trials.

 

 10   Now, would one consider, for example, kind of a

 

 11   super-DSMB not for the trial but for the drug that

 

 12   is being investigated by a commercial sponsor?

 

 13   Could a commercial sponsor, for example, if they

 

 14   are investigating drug X in, you know, 50 diseases

 

 15   in pediatrics, in geriatrics, and whatever, to have

 

 16   a coordinating center to look at this and then

 

 17   issue some type of report on these individual

 

 18   toxicities?

 

 19             Again, I understand exactly where Peter is

 

 20   coming from and the comments, having been there.

 

 21   You know, you get all this morass of information

 

 22   which is almost useless because nobody knows what

 

 23   to do about it and we are just generating paperwork

 

 24   with a pretense basically that we are doing

 

 25   something to further not only children but also

 

                                                               148

 

  1   adult clinical trials.

 

  2             Here again, you know, although we are

 

  3   talking about pediatrics, this does have obviously

 

  4   ramifications for adult medicine and adult clinical

 

  5   trials.  Although we may want to kind of say, well,

 

  6   some of the adult toxicities may not protect for

 

  7   what may go on into childhood toxicity, again, that

 

  8   is another level of clinical judgment and

 

  9   subjectivity that comes into play here.  Many of

 

 10   these drugs, especially with the IRBs, are not

 

 11   solely being looked at in children.  In many of

 

 12   your hospitals, since you practice exclusively in

 

 13   children's hospitals, that may be the case and your

 

 14   interest may be in that group, but for a garden

 

 15   variety IRB at a university hospital they may have

 

 16   ongoing studies in adults with breast cancer, colon

 

 17   cancer and pediatrics.  So, they need to look at

 

 18   this so it isn't that helpful sometimes to the

 

 19   larger IRBs, the university IRBs.

 

 20             DR. SANTANA:  Dr. Reynolds, you had a

 

 21   comment?

 

 22             DR. REYNOLDS:  I wanted to make it clear

 

 23   that the DSMB process is a little different in the

 

 24   pediatric setting.  You have one for your

 

 25   consortia, don't you, that look at all the studies

 

                                                               149

 

  1   that are going on and that are not specific to a

 

  2   drug.  But I think taking that in the context of

 

  3   what we are hearing from Ruth, and I hear this

 

  4   continually from a lot of people, the burden that

 

  5   is placed on the IRBs at the institutional level is

 

  6   substantial.

 

  7             Just taking a round number of 20

 

  8   institutions in your consortia, Peter, you have 20

 

  9   different IRBs looking at each one of these adverse

 

 10   events.  How many people are on each of those IRBs?

 

 11   Certainly, the total number far exceeds the number

 

 12   of patients on a study by an order of magnitude or

 

 13   two.  So, it is that process, yet we have a

 

 14   centralized DSMB process.  So the real central

 

 15   issue though comes down to the responsibility that

 

 16   the IRBs have under the regulations to be the

 

 17   ultimate and final arbitrator of whether or not

 

 18   this is going to be safe and appropriate for the

 

 19   patients in their institution.

 

 20             Somehow we need to use the word that I

 

 21   first learned in the context of Steve Hirschfeld,

 

 22   "harmonization."  I see it over and over again with

 

 23   the regulations you are harmonizing.  I think we

 

 24   need to somehow harmonize this process so that we

 

 25   can then decrease the workload for these poor

 

                                                               150

 

  1   people in the IRBs that, as you have heard, are

 

  2   volunteering their time and they are a precious

 

  3   resource that we could exhaust and then we wouldn't

 

  4   have anymore volunteers.

 

  5             DR. SANTANA:  I want to follow-up on a

 

  6   comment related to the previous issue of whether

 

  7   there should be another body that could help us

 

  8   review these things and probably give better

 

  9   knowledge to practicing oncologists.  You know, one

 

 10   of the concerns I always have about creating

 

 11   another body is that you don't destroy mass; it

 

 12   doesn't go away; you are just shifting it to

 

 13   another group.  If we do that, I think we run the

 

 14   same risk without clear guidance of what that group

 

 15   needs to be doing.  They are going to be getting

 

 16   the same paperwork we are getting now.  So, unless

 

 17   there is guidance at the first step, which is let's

 

 18   clearly define what we should be looking at and

 

 19   streamline that, it doesn't really matter where it

 

 20   goes to, whether it goes to an IRB, to a DSMB or to

 

 21   another group or another consortium.

 

 22             I think that may solve part of the problem

 

 23   but it is really shifting a little bit of the

 

 24   responsibility and what I want to get at is that we

 

 25   should probably encourage ourselves more to define

 

                                                               151

 

  1   the responsibility and the process rather than

 

  2   creating another group.  That is just a general

 

  3   comment.  It is not meant to be a criticism.  It is

 

  4   just something we need to think about.

 

  5             DR. KEEGAN:  Actually, going towards

 

  6   that--what you say, it doesn't help to create

 

  7   another group that is duplicating effort so it

 

  8   would only be effective if, in fact, the other

 

  9   groups then would agree to accept the information

 

 10   provided by the central group.  So, I think that

 

 11   has been the issue with central IRBs all along.

 

 12   While IRBs are crying out that they are

 

 13   overwhelmed, yet, they also refuse to defer that

 

 14   part of their responsibility to another group or to

 

 15   a central IRB.  Do you think that for a central

 

 16   pediatric IRB there is more willingness to do that,

 

 17   Malcolm?  I mean, are they willing to say, okay, we

 

 18   will allow somebody who is going to make an

 

 19   integrated analysis to do that and we will accept

 

 20   their judgment?

 

 21             DR. SMITH:  It will vary by institution.

 

 22   You know, the adult IRB has a facilitated review

 

 23   process and when a local IRB accepts the central

 

 24   IRB as the IRB of record, then the central IRB is

 

 25   responsible for the review of the adverse events

 

                                                               152

 

  1   relevant to that study.  In pediatrics, based on a

 

  2   survey that the Children's Oncology Group did,

 

  3   there is a high level of interest in a central

 

  4   pediatric IRB, both among PIs as well as among IRB

 

  5   chairs.  But when it comes to implementation, some

 

  6   institutions will accept it wholeheartedly and some

 

  7   won't.  But those who do will certainly be saving

 

  8   in terms of the effort expended on this.

 

  9             DR. SANTANA:  Peter, one last question.

 

 10             DR. ADAMSON:  I just wanted to follow-up

 

 11   on that.  So, it is not only the IRBs who are

 

 12   sometimes unwilling to give up the ability, it is

 

 13   the institution.  The institution more often than

 

 14   not will actually tell the IRB, you know what, we

 

 15   need an independent IRB; we are not going to accept

 

 16   it.  So, they may even take it out of the hands of

 

 17   the IRB as far as whether they are willing to or

 

 18   not.  So, IRBs are looking for ways to cut down

 

 19   their own work but it is not always coming to them.

 

 20             DR. SANTANA:  With that final

 

 21   comment--Ramzi, I will defer to you.

 

 22             DR. DAGHER:  Just very briefly, you seem

 

 23   to have identified a sense of challenges in terms

 

 24   of the filtering.  One is how to decide how

 

 25   relevant an adverse event is, and that is not

 

                                                               153

 

  1   really just specific to pediatric oncology or

 

  2   oncology, for that matter.  The second one, which

 

  3   Peter Adamson was trying to focus on, is how do you

 

  4   filter out the adult oncology experience or other

 

  5   experience that is submitted to you in terms of how

 

  6   relevant that is or isn't to the pediatric oncology

 

  7   setting.

 

  8             Now, you mentioned age and the nature of

 

  9   the adverse event.  Those are two potential

 

 10   criteria.  I am curious to know, and probably we

 

 11   will get into this more in answering the questions

 

 12   from Peter Adamson, Victor or others who have dealt

 

 13   with this, what criteria do you use in making

 

 14   decisions about filtering the adult oncology

 

 15   reported events and deciding how relevant they are

 

 16   to your specific studies?

 

 17             DR. SANTANA:  I think with that question

 

 18   we will go ahead and try to address the questions

 

 19   for the committee because I think we will cover

 

 20   that.

 

 21             DR. PRZEPIORKA:  Can I just ask one more

 

 22   question?

 

 23             DR. SANTANA:  Yes, Donna?

 

 24             DR. PRZEPIORKA:  You had indicated that,

 

 25   if I recall, your institution does not take

 

                                                               154

 

  1   patients off protocol so that you get long-term

 

  2   follow-up.  I was wondering if you thought that was

 

  3   appropriate for everybody to be doing in the

 

  4   pediatric population or if there is some time

 

  5   limit, like by age 35 we are not going to look

 

  6   anymore, or something like that?

 

  7             DR. SANTANA:  Well, if we are conducting

 

  8   active research on those patients, those patients

 

  9   would come off their primary therapeutic protocol

 

 10   and get enrolled on a non-therapeutic protocol,

 

 11   which is an umbrella protocol we have for long-term

 

 12   follow-up.  So, they would still be research

 

 13   participants and we are collecting data on

 

 14   long-term effects, survival and things like that.

 

 15   So, the patient would come off the primary

 

 16   therapeutic protocol once they are transitioned

 

 17   into the long-term follow-up protocol on which

 

 18   research is being conducted.  So, those active

 

 19   protocols will not show up in the reporter but the

 

 20   long-term follow-up will show up in the reporter

 

 21   for that patient.

 

 22                     Questions for Discussion

 

 23             Let's go ahead and try to address the

 

 24   questions that we have before us.  Just for the

 

 25   purpose of the minutes and the documents, I will go

 

                                                               155

 

  1   ahead and read the questions to the committee, the

 

  2   introduction, and then we will take one question at

 

  3   a time.

 

  4             The tolerance for risk in cancer

 

  5   therapeutics is different than for most other

 

  6   medical therapies.  It is also recognized that

 

  7   children are a particularly vulnerable population

 

  8   and regulations and procedures have been

 

  9   implemented to provide protection to children

 

 10   participating in clinical research.  The following

 

 11   questions relate to the setting of children with

 

 12   cancer participating in clinical trials.

 

 13             Under the heading of "principles" the

 

 14   question is, what are the principles that should be

 

 15   addressed in safety monitoring of clinical studies

 

 16   that enroll children with cancer?  If the

 

 17   principles are adequately stated in existing

 

 18   documents, statutes or regulations, please identify

 

 19   the relevant documents and sections.

 

 20             Barry or Malcolm, from the NCI

 

 21   perspective, do you have any comments on existing

 

 22   regulations or documents that we could reference

 

 23   to?

 

 24             DR. ANDERSON:  In terms of the DSMBs, the

 

 25   composition of DSMBs, that sort of information is

 

                                                               156

 

  1   provided in OHRP.  In terms of the frequency of

 

  2   monitoring and the exact nature of the monitoring,

 

  3   what is monitored which is part of the discussion

 

  4   we had, I don't know that that is laid out as

 

  5   clearly.  We have guidelines that we work with at

 

  6   CTEP and NCI but I don't know that that is in

 

  7   regulatory form at all.

 

  8             DR. SMITH:  Yes, there is the overall

 

  9   policy on data monitoring.  That is really not very

 

 10   prescriptive in terms of here is what you have to

 

 11   review; here is how often you have to look at it;

 

 12   and here is, you know, who should be looking at it.

 

 13   It says you need to have a plan but it is not very

 

 14   prescriptive in terms of what the plan is.  Each of

 

 15   the institutions has their own data and safety

 

 16   monitoring plans, particularly for Phase III

 

 17   trials, and those tend to be more prescriptive and

 

 18   detailed in terms of what is happening.  But in

 

 19   terms of early phase trials, you know, I am not

 

 20   aware of kind of NIH-generated documents that

 

 21   provide detail about what, how, when and where this

 

 22   needs to be done.

 

 23             DR. SANTANA:  Go ahead, Barry.

 

 24             DR. ANDERSON:  And having been on the

 

 25   panel of people who looked at the cancer center

 

                                                               157

 

  1   data and safety monitoring plans that they had to

 

  2   submit, previously I think a lot of people would

 

  3   recognize that for early phase studies it was the

 

  4   investigator and their research nurse that looked

 

  5   over the data with the most frequency.  A lot of

 

  6   times I think there was not a lot of oversight from

 

  7   outside of that small group.  It was clear from

 

  8   looking at the different cancer centers that there

 

  9   is a huge spectrum of what in reality they were

 

 10   doing and when you told them, you know, you need to

 

 11   formalize this what they presented us with what

 

 12   they thought were acceptable approaches.  From our

 

 13   point of view, we had these essential elements to

 

 14   work from but they are very general and it took us

 

 15   a while to kind of gear up to say here is exactly

 

 16   what we think--well, not exactly but here is a

 

 17   range of possibilities that are acceptable as an

 

 18   approach, and I think it does vary by the type of

 

 19   study that is actually being considered.  That was

 

 20   one of the criteria, for Phase I studies we would

 

 21   do this; for pilots, this.  For Phase II and Phase

 

 22   III there were different levels of monitoring that

 

 23   seemed to be appropriate for each of those, both in

 

 24   terms of the type of monitoring and the frequency

 

 25   of kind of review of the data and that type of

 

                                                               158

 

  1   thing.

 

  2             DR. SANTANA:  As a follow-up to that, in

 

  3   the non-NCI cancer center umbrella, all the other

 

  4   groups that NCI supports like the consortia, are

 

  5   there also specific requirements for DSMB plans for

 

  6   those consortia?

 

  7             DR. SMITH:  The overall NIH requirements

 

  8   apply to all NIH-sponsored research.  Again, those

 

  9   require a data monitoring plan, not a particular

 

 10   form that that plan has to take for implementation.

 

 11   I guess one question here is does FDA want kind of

 

 12   the form and the details, or is it a question of

 

 13   principles, you know, whatever the plan is, it

 

 14   should adhere to these principles?

 

 15             DR. SANTANA:  I think with that comment, I

 

 16   will ask Eric--are you still on the line?

 

 17             DR. KODISH:  I am here.

 

 18             DR. SANTANA:  Eric, can you comment on

 

 19   that in trying to address the issue of global

 

 20   principles, other than specific detail?

 

 21             DR. KODISH:  I would opt for flexibility--

 

 22             DR. SANTANA:  Eric, can you speak just a

 

 23   little bit louder, please?

 

 24             DR. KODISH:  Yes.  I would argue for

 

 25   flexibility.  I think that the different contexts

 

                                                               159

 

  1   of the particular clinical trials involving

 

  2   children with cancer that we are talking about

 

  3   would dictate that it makes more sense to allow a

 

  4   plan based on principles, such as beneficence or

 

  5   such as filtering serious adverse events compared

 

  6   to those that are not as impactful, and I wouldn't

 

  7   try to prescribe the format so much.  That would

 

  8   lead to bureaucratization that could actually

 

  9   paradoxically harm the ethical importance of

 

 10   research.

 

 11             DR. HIRSCHFELD:  I would like a

 

 12   clarification from Dr. Kodish.  So, would you then

 

 13   say that the principles of, let's say, beneficence

 

 14   and respect contained in the Belmont report and the

 

 15   principles that are annunciated in the ICH

 

 16   documents, for instance particularly the one that

 

 17   applies to pediatric research, E11, are a

 

 18   sufficient statement of the principles?

 

 19             DR. KODISH:  I would.

 

 20             DR. HIRSCHFELD:  I think we can move on.

 

 21             DR. SANTANA:  Before we get to that

 

 22   question though, because I want to make sure that

 

 23   we cover the whole loop of this point, do

 

 24   pharmaceutical sponsors in their DSMB plans have

 

 25   any specific requirements for pediatrics, or are

 

                                                               160

 

  1   pediatrics dealt with in monitoring plans as the

 

  2   greater universe of adults?  Or has that ever been

 

  3   discussed, that they should develop specific plans

 

  4   for pediatrics?

 

  5             DR. GRILLO-LOPEZ:  Not to my knowledge

 

  6   but, again, I may not be the best person to address

 

  7   that.  On the other hand, I would like to comment

 

  8   on the subject of DSMBs because I would not like

 

  9   the FDA to come away from this meeting thinking

 

 10   that there is an endorsement for DSMBs to be

 

 11   required and/or regulated in any way, shape or

 

 12   form.  I think that there may be a need for some

 

 13   consensus agreement at the level of professional

 

 14   societies, the NIH and so on, on how different

 

 15   DSMBs might be constructed and when they may or may

 

 16   not be required, but allowing for the flexibility

 

 17   that several around the table have mentioned.

 

 18             DR. SANTANA:  That was my interpretation

 

 19   of the discussion too.  I don't think there was any

 

 20   endorsement from this group that we should be

 

 21   moving towards a model DSMB to solve some of the

 

 22   problems.

 

 23             DR. GRILLO-LOPEZ:  I see Dr. Pazdur

 

 24   agreeing with that and I am glad to see that.

 

 25             DR. SANTANA:  I want to clarify that that

 

                                                               161

 

  1   was my interpretation too.  That is not what I

 

  2   think the comment was all about.  Eric, did you

 

  3   want to add anything else?  I am sorry, I think I

 

  4   interrupted you.  No?

 

  5             DR. KODISH:  No, that is fine.

 

  6             DR. SANTANA:  So, we will move on then

 

  7   from question one--oh, Malcolm, I am sorry.

 

  8             DR. SMITH:  I think those are good

 

  9   principles but I think one can get a bit more

 

 10   detailed without being prescriptive in terms of

 

 11   what the principles of study monitoring should be.

 

 12   For example, the principle that study monitoring

 

 13   should be performed by experienced experts and that

 

 14   that review should be timely, and that whatever the

 

 15   system is, it should have those characteristics.

 

 16   And, study monitoring should be done in a way so

 

 17   that conflict of interest issues are addressed, and

 

 18   that study monitoring in whatever setting,

 

 19   especially in Phase III settings but even in Phase

 

 20   II settings and others is done in such a way that

 

 21   the integrity of the study and the confidentiality

 

 22   of data, when that is important, are addressed.

 

 23   So, I think there are principles of ethics that we

 

 24   need to adhere to and there are principles of

 

 25   monitoring that I think need to be clearly stated

 

                                                               162

 

  1   so that you can benchmark how you are addressing

 

  2   those basic principles of monitoring.

 

  3             DR. GRILLO-LOPEZ:  If I may, most of that

 

  4   is already covered in GCP and in other regulations.

 

  5             DR. SANTANA:  So noted.  I would only add

 

  6   to that that I think an essential element to that

 

  7   is this concept that I advocate, that there has to

 

  8   be an open communication with the research team,

 

  9   that monitoring doesn't occur in isolation from the

 

 10   actual research team that is conducing the study.

 

 11   I am not implying that the research team should be

 

 12   doing their own monitoring.  It shouldn't be

 

 13   interpreted that way but the research team should

 

 14   be integral to that process.  Dr. Reynolds?

 

 15             DR. REYNOLDS:  Malcolm, could I just ask

 

 16   you to elaborate on what the role of that DSMB is

 

 17   in the conflict of interest monitoring that you

 

 18   were talking about?

 

 19             DR. SMITH:  What the role of the DSMB is

 

 20   in conflict of interest?

 

 21             DR. REYNOLDS:  Did I hear you correctly?

 

 22   Were you saying that they are really involved in

 

 23   that role?

 

 24             DR. SMITH:  No, that the monitoring is

 

 25   done in such a way that conflict of interest issues

 

                                                               163

 

  1   are addressed.

 

  2             DR. REYNOLDS:  In other words, that the

 

  3   DSMB is a separate body and is not subject to

 

  4   conflict of interest.  That is what you are saying?

 

  5             DR. SMITH:  Well, that is one way of

 

  6   addressing it but not the only way of addressing

 

  7   conflict of interest issues, but that those issues

 

  8   are considered, both the financial and intellectual

 

  9   conflict of interest that may lead people to ask

 

 10   questions about decisions that are made.

 

 11             DR. KODISH:  This is Eric, in Cleveland.

 

 12   Another way of saying that I think is that

 

 13   transparency is an important principle, perhaps the

 

 14   idea that whatever the monitoring plan is that the

 

 15   appearance of the fox watching the henhouse won't

 

 16   be something that people can interpret as having

 

 17   gone on.

 

 18             DR. SMITH:  The Pediatric Phase I

 

 19   Consortium and the Pediatric Brain Tumor Consortium

 

 20   both have independent data monitoring committees,

 

 21   and these are early phase clinical trials.  They

 

 22   are not so much looking over the day to day

 

 23   activities of the consortium and every independent

 

 24   decision, but at intervals they are looking at the

 

 25   overall conduct of how these studies are being done

 

                                                               164

 

  1   and are an independent body that tries to address

 

  2   some of the conflict of interest issues, in this

 

  3   case particularly intellectual conflict or kind of

 

  4   ownership conflict issues, and to make sure that

 

  5   the research team is appropriately making decisions

 

  6   as they are conducting the studies.  They are there

 

  7   to provide guidance if difficult decisions arise

 

  8   about what their advice would be about how to

 

  9   address these difficult decisions.

 

 10             DR. SANTANA:  If there is no further

 

 11   comment on that we will move on to number two.  The

 

 12   next series of questions are more related to

 

 13   reality and practice.  Recognizing that particular

 

 14   populations, disease settings, and products may

 

 15   have specific requirements, what general parameters

 

 16   should be monitored for safety in all clinical

 

 17   studies?

 

 18             DR. HIRSCHFELD:  I should say all

 

 19   pediatric oncology clinical studies, just to be

 

 20   clear about that.

 

 21             DR. SANTANA:  So noted.  Peter?

 

 22             DR. ADAMSON:  I will take a stab at that.

 

 23   I think it very much depends on the phase of the

 

 24   study.  In pediatrics I think we have some

 

 25   advantages in that for Phase III studies there is

 

                                                               165

 

  1   probably a general standard of care that we follow

 

  2   whether a child is or is not on study as far as

 

  3   frequency of monitoring.  I would say that that

 

  4   would probably be the minimum threshold for Phase

 

  5   III studies.

 

  6             As one marches down from Phase III to

 

  7   Phase II and Phase I, I think this is where Phase I

 

  8   cancer is different than Phase I "the rest of the

 

  9   world" because we conduct the Phase I studies in

 

 10   patients with the disease.  So, I don't think you

 

 11   can layer the same level of monitoring as you do in

 

 12   other studies where volunteers are locked away for

 

 13   two weeks and are plugged into every known device

 

 14   to see what happens.  We can't do that.

 

 15             I think we need to look at preclinical

 

 16   data as far as what potential toxicities are, and

 

 17   in children we have the advantage of looking at the

 

 18   initial adult Phase I experience to see what the

 

 19   relevant additional monitoring might be required.

 

 20   We shouldn't be getting PFTs, echoes, EKGs, stress

 

 21   tests, all the way down the line if, in fact, that

 

 22   is not relevant to a particular drug.  So, I think

 

 23   we have the advantage of looking at the Phase I

 

 24   adult experience.  Then, we always have to balance

 

 25   the level of monitoring, recognizing that these are

 

                                                               166

 

  1   patient volunteers and not normal volunteers as far

 

  2   as trying to strike a balance.

 

  3             DR. SANTANA:  Pamela?

 

  4             MS. HAYLOCK:  I am not sure how relevant

 

  5   this is but you keep talking about monitoring and I

 

  6   think a lot of this has to do with expanding the

 

  7   definition of safety and monitoring in regards to

 

  8   concepts that involve long-term and late survivors.

 

  9   Your institution is maybe somewhat unique in having

 

 10   long-term survivorship programs, but not all places

 

 11   which do pediatric research have such things, and

 

 12   now we are ending up with adult survivors of

 

 13   childhood cancers who are 10, 20, maybe 3 or 4

 

 14   decades out who are experiencing surprise long-term

 

 15   related effects and I think somehow the parameter

 

 16   of safety and monitoring needs to be expanded.  I

 

 17   don't know how to do that but I think the late

 

 18   effects need to be a consideration.

 

 19             DR. SANTANA:  Actually, cooperative groups

 

 20   and other pediatric consortia are addressing that.

 

 21   I mean, I think there is a big effort at the

 

 22   cooperative group level to look at long-term

 

 23   survivor issues in pediatric oncology patients.

 

 24   Obviously, it is in different stages but I think we

 

 25   all recognize as pediatric oncologists that that is

 

                                                               167

 

  1   an issue, and I think it is being addressed at

 

  2   different levels.  Malcolm and then Donna?

 

  3             DR. SMITH:  It is a critical issue.  The

 

  4   challenge with it is that you are looking 10, 20

 

  5   and 30 years up the road so the infrastructures,

 

  6   like the children's hospitals around the table,

 

  7   need to reach out to a lot of other institutions

 

  8   and to the survivors in order for that work to be

 

  9   done.  So, there are different ways that the

 

 10   Children's Oncology Group, the childhood cancer

 

 11   survivor study are trying to address that, and it

 

 12   is recognized as an important issue that we have to

 

 13   address.

 

 14             DR. SANTANA:  Donna?

 

 15             DR. PRZEPIORKA:  I just wanted to ask, the

 

 16   organized groups and the major institutions clearly

 

 17   have a plan but what about industry?  I mean,

 

 18   industry does do pediatric trials.  What sort of

 

 19   guidance do you give to them, and what is the basis

 

 20   for that guidance?  I mean, what has come out of

 

 21   the St. Jude experience monitoring long-term

 

 22   survival in their patients, and is it really worth

 

 23   mandating that the pharmaceutical

 

 24   industry-sponsored trials do long-term follow-up?

 

 25             DR. SANTANA:  I think the issue of

 

                                                               168

 

  1   long-term survivorship follow-up and data needs to

 

  2   be considered by the pharmaceutical industry when

 

  3   they are developing a drug in terms of the

 

  4   long-term issues that may be particular to that

 

  5   drug.  The problem comes there that the sponsors

 

  6   themselves are limited to a period of time in terms

 

  7   of when they are doing the project with you.  Once

 

  8   the project is over, then the responsibility of

 

  9   monitoring patients long term becomes the

 

 10   responsibility of the treating institution.  So up

 

 11   front, at least in my experience in all the studies

 

 12   that I have participated in with pharmaceutical

 

 13   industry, I have never really seen, within the

 

 14   context of the protocol, any plan for long-term

 

 15   issues that may arise as a result of follow-up of

 

 16   these patients.  Once a study is done, it is done

 

 17   and then it becomes the responsibility of the

 

 18   treating institution to decide what they are going

 

 19   to look for, how it is collected and how it is

 

 20   analyzed.  So, there is a little bit of a dis-link

 

 21   there in that we have never really required or

 

 22   asked pharmaceutical industry to address that in

 

 23   the context of the front-line trial that is being

 

 24   developed.  Peter?

 

 25             DR. ADAMSON:  Again, pediatrics in this

 

                                                               169

 

  1   respect differs from adults because where you

 

  2   really get the long-term ability to look at late

 

  3   effects is in or following Phase III.  I am not

 

  4   aware of any industry-sponsored Phase III studies

 

  5   in pediatric oncology.  They are almost universally

 

  6   done within the cooperative groups.  There are

 

  7   industry-sponsored Phase I and Phase II studies,

 

  8   without question.  I think our ability to really

 

  9   ask late effects questions in that population is

 

 10   severely limited so it really becomes the burden of

 

 11   the NCI and the cooperative groups when conducting

 

 12   Phase III trials and, as Malcolm said, there is a

 

 13   whole separate late effects effort.  So, I don't

 

 14   think it is something that realistically we can

 

 15   burden industry with because of the likelihood of

 

 16   getting that data in a Phase I or Phase II study.

 

 17   If the environment were to change and we would

 

 18   dream that industry would support a Phase III

 

 19   randomized study in children, then I think we would

 

 20   have to look at the willingness to look for

 

 21   long-term effects.

 

 22             DR. SANTANA:  I will correct myself.  I am

 

 23   aware of one study that I have seen, which is an

 

 24   antibiotic study that is actually being sponsored

 

 25   by industry, looking at some issues of long-term

 

                                                               170

 

  1   effects of the use of that antibiotic in a

 

  2   pediatric population.  It is a very long-term

 

  3   study.  It is a very costly study too.  So, I am

 

  4   aware of that example that came to mind as I was

 

  5   hearing the discussion but that is kind of unique.

 

  6             DR. ADAMSON:  And it is not

 

  7   anti-neoplastic therapy.

 

  8             DR. SANTANA:  No, it is not.  It is an

 

  9   antibiotic study.  Any other guidance we can give

 

 10   you on this question, Dr. Hirschfeld or Dr. Pazdur?

 

 11   Yes?

 

 12             MS. HOFFMAN:  I think integral to

 

 13   monitoring safety also in terms of when a child is

 

 14   on treatment is also monitoring participation and

 

 15   entering into the study, and I think we need to

 

 16   monitor informed consents and parents'

 

 17   comprehension of randomization, especially in Phase

 

 18   I studies.  Are they really understanding what they

 

 19   are getting into?  Also, monitoring waiver of

 

 20   consents because I think there is potential

 

 21   conflict of interest there.  The waivers that are

 

 22   coming to the IRB are coming from the PI who is

 

 23   often the clinician as well of the child and,

 

 24   again, there could be conflict there.  So, again, I

 

 25   think it is a safety monitoring issue.

 

                                                               171

 

  1             DR. SANTANA:  I will try and summarize

 

  2   what I heard as committee discussion of this

 

  3   question.  I think the committee was pointing out

 

  4   that in a certain way we have a little bit of an

 

  5   advantage in that there may be some adult data

 

  6   before pediatric studies are initiated, and a lot

 

  7   of the safety issues and monitoring that we would

 

  8   want to do in pediatrics have to be put in the

 

  9   context of what data already exist in the adult

 

 10   population that has received those drugs, but also

 

 11   considering that there may be specific niches that

 

 12   pediatrics would provide that we have to look for

 

 13   that may not have been identified in the adults.  I

 

 14   heard that comment.

 

 15             I heard the other comment, that it has to

 

 16   be developmentally phase dependent in terms of what

 

 17   type of study you are talking about, that the issue

 

 18   of safety monitoring is very different in a Phase

 

 19   III trial than it would be in a Phase I, and that

 

 20   there are different mechanisms of reaching those.

 

 21   In a Phase I it may be more the research team, the

 

 22   consortium group continuously looking at that data

 

 23   and making safety judgments, whereas in a Phase III

 

 24   it may be a DSMB or may be other regulatory bodies

 

 25   that can define what safety issues need to be

 

                                                               172

 

  1   looked at and how they are evaluated.  I heard that

 

  2   comment.

 

  3             I think the third comment I heard was

 

  4   about this issue of paying some attention to the

 

  5   initial enrollment of patients on studies,

 

  6   pediatric oncology studies, and how we can more

 

  7   effectively not only monitor their involvement but

 

  8   get some degree of understanding of what people

 

  9   really are hearing and their assessment of risk and

 

 10   what they think they are participating in.

 

 11             Those are the three comments I kind of

 

 12   heard around the table.  Susan?

 

 13             DR. WEINER:  I have one more, which is

 

 14   that I really haven't heard any discussion this

 

 15   morning of the notion of safety in trials of

 

 16   biologics where toxicity may not be what you are

 

 17   looking for in a Phase I trial, and it is not clear

 

 18   to me how we might approach that in this context.

 

 19             DR. SANTANA:  That is a good point.

 

 20             DR. HIRSCHFELD:  Noted.

 

 21             DR. KEEGAN:  I think you also should

 

 22   consider that it may be toxicity, it may be other

 

 23   examples but one shouldn't exclude the fact that

 

 24   toxicity could also be a component even in biologic

 

 25   trials.

 

                                                               173

 

  1             DR. GRILLO-LOPEZ:  I was just going to

 

  2   reinforce what Dr. Keegan said.  You know, in the

 

  3   past two years having developed two biologics, they

 

  4   were both associated with some toxicities that were

 

  5   important.  So, one has to be careful, going into

 

  6   the development of a biologic, not to think that

 

  7   there might be fewer, lesser toxicities.  So, one

 

  8   really has to do the same monitoring that one would

 

  9   do for a chemotherapeutic agent until one is sure

 

 10   of what the toxicity profile is for that particular

 

 11   biologic.

 

 12             DR. WEINER:  Or expand those definitions.

 

 13             DR. HIRSCHFELD:  I think we all agree that

 

 14   the spectrum and the severity may vary but there is

 

 15   no intervention that is risk free.

 

 16             DR. KEEGAN:  Yes, I think the principles

 

 17   Dr. Adamson mentioned were, you know, looking at

 

 18   the nonclinical and adult data to guide what would

 

 19   be used for biologics and even for a lot of

 

 20   traditional drugs, you know, small chemical drugs

 

 21   that are targeted in some way.

 

 22             DR. SANTANA:  Yes, I want to add that

 

 23   there was another point that was made as a general

 

 24   consensus point as advice to the agency that had to

 

 25   do with the issue of neurocognitive development,

 

                                                               174

 

  1   and that that may be a particular issue in terms of

 

  2   safety that should be addressed in safety

 

  3   parameters in pediatric oncology trials.  In

 

  4   contrast to some of the things that we could

 

  5   capture from adult trials, that is particularly

 

  6   unique to pediatric trials and we should pay some

 

  7   attention to it.  Donna?

 

  8             DR. PRZEPIORKA:  Actually, just to

 

  9   follow-up on that, the one other piece of

 

 10   information that I think is very easy to obtain and

 

 11   to analyze is growth.

 

 12             DR. SANTANA:  Any further comments on that

 

 13   question?  If not, we will move on to the next

 

 14   question.  Based on the response to the previous

 

 15   question, how often should the parameters be

 

 16   monitored?

 

 17             Here I would say I think we need to be

 

 18   careful.  We don't want to get into a prescription

 

 19   plan that everybody has to do kind of in the same

 

 20   way in terms of what things get monitored, at what

 

 21   particular time intervals and how often.  I think

 

 22   the idea that I proposed when we looked at our plan

 

 23   at our institution is that it is phase dependent.

 

 24   Once again we go back to the phase issue of the

 

 25   type of study that you are conducting.  So,

 

                                                               175

 

  1   particular Phase I studies may be monitored more

 

  2   frequently than other Phase I studies.  Maybe some

 

  3   biologic studies, gene transfer studies that are

 

  4   Phase I need to be monitored more frequently than

 

  5   an oncology Phase I study.

 

  6             The point I want to make is that although

 

  7   it is phase dependent, I think also in the formula

 

  8   has to be included the specific agent that you are

 

  9   testing in that phase in order to decide how often

 

 10   you are going to monitor it.  Peter?

 

 11             DR. ADAMSON:  Yes, I would echo that.

 

 12   Again, going back to the adult experience, it gives

 

 13   us an advantage as far as what to expect and when

 

 14   to expect it.  But the other thing that we

 

 15   sometimes err on is that we have to look at our own

 

 16   definitions of toxicity and what we consider either

 

 17   serious or dose-limiting because when you look at

 

 18   those definitions, you then look at how frequently

 

 19   you are monitoring and you realize you will never

 

 20   be able to meet those definitions.  So, as I said,

 

 21   perhaps a simple starting place if you want to get

 

 22   some idea of what the spectrum is, there a number

 

 23   of cooperative groups or a number of single

 

 24   institutions that conduct this and my guess is you

 

 25   will find a common thread in the backbone of those

 

                                                               176

 

  1   that apply across the board for Phase I and a

 

  2   different set for Phase II and then it becomes very

 

  3   agent dependent beyond that.

 

  4             DR. PAZDUR:  I have a question as far as

 

  5   the toxicity criteria for children, are there any

 

  6   differences between that and what we use for

 

  7   adults, other than perhaps physiological

 

  8   differences that may exist with growth parameters?

 

  9   What I am after is some of our adult toxicity

 

 10   criteria have some subjective elements as far as

 

 11   elements of daily activity, fatigue, etc., and how

 

 12   do you figure that into toxicity assessments with

 

 13   children?  Or, do they have difficulty in assessing

 

 14   some of these toxicities in children?  You know,

 

 15   for some of our activities for adults neurotoxicity

 

 16   might be difficulty in buttoning your shirt or in

 

 17   adult activities of daily living in a sense.

 

 18             DR. SANTANA:  Alice, it looks like you

 

 19   wanted to respond to that.

 

 20             MS. ETTINGER:  Well, I think we all

 

 21   understand that for kids we have to look at them at

 

 22   an age appropriate level and many times that would

 

 23   be school attendance, how they are functioning in

 

 24   school, certainly measurements of that sort.  I

 

 25   think in terms of fatigue, we are way behind in

 

                                                               177

 

  1   measuring the actual fatigue level that we may be

 

  2   seeing in children, not only little ones but

 

  3   certainly as they grow up.  Often in filling out

 

  4   the forms for doing the criteria, I feel that there

 

  5   may actually need to be other criteria that we look

 

  6   at and that we measure for children.

 

  7             DR. SANTANA:  It is a good point.  The

 

  8   issue with those criteria is that as yet they

 

  9   haven't been validated so it is very hard to apply

 

 10   them across studies but there is actually a lot of

 

 11   research going on in that field that, hopefully, in

 

 12   the next few years will give us some guidance.  But

 

 13   that is the problem, those criteria are soft and

 

 14   they haven't been validated so it is very hard to

 

 15   apply them.  So, in oncology we kind of rely on the

 

 16   standard toxicity criteria that was developed by

 

 17   NCI, etc., in terms of what we look for and how we

 

 18   code it.

 

 19             DR. HIRSCHFELD:  I will just add to that.

 

 20   There have been questions raised about having some

 

 21   pediatric specific scales, but it was the absence,

 

 22   as Dr. Santana pointed out, of having validated

 

 23   assessments that has precluded from formally

 

 24   incorporating those.  So, that is an area that

 

 25   still remains under discussion and has had some

 

                                                               178

 

  1   interest for some years.

 

  2             DR. ANDERSON:  And in the current version

 

  3   of the CTC, the updated version that just came out,

 

  4   where possible, all distinctions between pediatric

 

  5   and adult criteria were eliminated because

 

  6   basically we generalize the grading.  I can't

 

  7   remember exactly what word you used, Dr. Pazdur, in

 

  8   terms of the degree of toxicity.  You know, just

 

  9   having treated patients with different pediatric

 

 10   cancers and actually having heard from people who

 

 11   are trying to set up studies with certain

 

 12   dose-limiting toxicities, in pediatrics a lot of

 

 13   times I think the dose-limiting toxicities that we

 

 14   accept are greater than are accepted in adults.

 

 15   They will stop an adult trial or they will change

 

 16   an individual adult's treatment much sooner than we

 

 17   do in pediatric oncology and I don't know that we

 

 18   have different measurements of toxicity but we

 

 19   would move a grade further perhaps, or half a grade

 

 20   further in terms of maybe the duration of the

 

 21   tolerance of a toxicity than happens in medical

 

 22   oncology.

 

 23             DR. SANTANA:  And those are usually

 

 24   specifically defined within the context of a

 

 25   protocol.  So, for some studies we would accept up

 

                                                               179

 

  1   to grade X and in others we wouldn't.  So, I think

 

  2   there is a lot of variability and it is really

 

  3   driven by the protocol and the question you are

 

  4   trying to answer and what you know about that drug

 

  5   beforehand.

 

  6             The next question is based on the response

 

  7   to the previous question, who should do the

 

  8   monitoring?  Is it adequate to have the personnel

 

  9   involved in the study be responsible for safety

 

 10   monitoring?  Susan?

 

 11             DR. WEINER:  The issue of the conflict of

 

 12   role between the investigator and the treating

 

 13   physician is something that has been discussed over

 

 14   the past few years in a variety of contexts.  I

 

 15   think that applying that notion to this, it becomes

 

 16   obvious that such a team is insufficient.

 

 17             DR. SANTANA:  Peter?

 

 18             DR. ADAMSON:  I guess I would disagree

 

 19   with that to an extent.  It very much depends I

 

 20   think on the phase of the study, and the number of

 

 21   children who are at risk, and what the goals of the

 

 22   study are.  From a practical standpoint, for a

 

 23   Phase I where the study is a real-time study that

 

 24   is the role of the study team.  They are making a

 

 25   decision on a patient to patient basis.  Having an

 

                                                               180

 

  1   additional layer of oversight to make sure the

 

  2   study team is meeting its obligations I think is

 

  3   helpful and is important but, from a practical

 

  4   standpoint, you can't convene a data safety

 

  5   monitoring board with every dose escalation step;

 

  6   you never would end up conducting the study.

 

  7             Having said that, it is important to keep

 

  8   in mind that the goal of a Phase I study is to

 

  9   recommend a dose and so the study is going to be

 

 10   successful really no matter where you stop as far

 

 11   as an investigator conflict of interest.  I mean,

 

 12   they will meet their study endpoint.  Having said

 

 13   that, when you come to a Phase III, you really do

 

 14   need additional layers of monitoring because then

 

 15   you really want to prove is this drug effective and

 

 16   there is a lot riding on the outcome of that study.

 

 17             So, the level of monitoring I think very

 

 18   much depends on what the phase of the study is.

 

 19   But I think, without question, you need to know

 

 20   what the data safety monitoring plan is.  I mean,

 

 21   investigators need to be very clear and very

 

 22   specific up front about how this study is going to

 

 23   be monitored.  I will come back to what Eric Kodish

 

 24   had said earlier, you need to have some flexibility

 

 25   as far as what the level of monitoring is and who

 

                                                               181

 

  1   does it.  If it is a cytotoxic and there is a lot

 

  2   of experience in developing the cytotoxic, that may

 

  3   lead to one level.  If it is an entirely new

 

  4   modality of treatment being put forward, you may

 

  5   want to consider another layer of monitoring.  So,

 

  6   there has to be some flexibility within the system.

 

  7             DR. SANTANA:  I would echo what Peter

 

  8   said.  I think it is a graded system and it depends

 

  9   on the type of study you are doing and what

 

 10   elements are being monitored.  For example, if you

 

 11   want to get into the nitty-gritty details of

 

 12   monitoring enrollment and informed consent, I think

 

 13   that has to be independent of the research team.

 

 14   There is no other way you could do that; it has to

 

 15   be a separate monitoring group that does that,

 

 16   whether it is the protocol office or another group

 

 17   of people.  But in a Phase I study if the central

 

 18   question is the toxicity, that should be monitored

 

 19   by the study team because that is what is going to

 

 20   define how the study progresses.  Then you may have

 

 21   intervals in which that data is shared with a

 

 22   central Phase I group, etc., etc.

 

 23             Whereas, in a Phase III study you are in a

 

 24   completely opposite direction.  For a Phase III

 

 25   study most of the elements for safety that you want

 

                                                               182

 

  1   to monitor have to be done independent of the

 

  2   investigator.  They are large group studies with

 

  3   data collection.  There may be some safety issues

 

  4   that have to be reported to the safety data

 

  5   monitoring boards so you have to use those

 

  6   resources.

 

  7             So, I don't see it as black and white.  I

 

  8   see it as a graded system in which the elements

 

  9   that are going to be monitored, the safety and how

 

 10   that is done may incorporate different groups and

 

 11   you just have to find the right fit for the study

 

 12   that you are considering.  I hate to put it in

 

 13   black and white; it won't work if it is black and

 

 14   white.  I think the beauty of some of the stuff

 

 15   that Peter mentioned in terms of what the Phase I

 

 16   and the COG Consortium is doing is that they are

 

 17   doing it in real time.  I mean, they are looking at

 

 18   that week by week, maybe two weeks or however

 

 19   often, so they have the advantage of doing that in

 

 20   real time so that they can intervene if they have

 

 21   to.  Whereas, I think that would be impossible to

 

 22   do in a Phase III study.  You just couldn't get

 

 23   people to do that.  Dr. Reynolds?

 

 24             DR. REYNOLDS:  Peter, acknowledging the

 

 25   challenges you put forth that a data safety

 

                                                               183

 

  1   monitoring board in a Phase I study--that it is not

 

  2   practical for them to convene and review, I think

 

  3   we should acknowledge that there are some

 

  4   significant advantages to having such a board for

 

  5   the day to day people that are monitoring to go to

 

  6   with questions about study design amendments that

 

  7   might make it more acceptable from a safety

 

  8   standpoint, and having that group that is external

 

  9   to the people who are actually conducting the

 

 10   study.  It is a small world in pediatrics, so

 

 11   having that separated out, at least from the NANT

 

 12   perspective, is a great advantage in being able to

 

 13   bounce things off these people externally.

 

 14             DR. SANTANA:  Dr. Smith?

 

 15             DR. SMITH:  We talked about NANT trials,

 

 16   COG trials and we are very restricted to that.

 

 17   Would there be a separate answer for

 

 18   industry-sponsored Phase I/Phase II trials?  Is

 

 19   that a different situation?

 

 20             DR. SANTANA:  Usually in Phase I

 

 21   industry-sponsored trials, at least the ones I am

 

 22   familiar with, there is a research team that is

 

 23   identified.  It is usually the PI at various

 

 24   institutions; it is a medical officer or monitor

 

 25   from the pharmaceutical company or contact person. 

 

                                                               184

 

  1   I think the same functional principle can be

 

  2   applied, that that research team should communicate

 

  3   frequently and often enough as the study is being

 

  4   conducted to make ongoing decisions about the

 

  5   safety of the study.  So, I think that may already

 

  6   be happening.  We just don't know about it.  If it

 

  7   is not happening, we should probably extend those

 

  8   things that we are doing in some of these consortia

 

  9   to those.  I think they are practical and they

 

 10   don't require a lot more work.

 

 11             DR. GRILLO-LOPEZ:  If I may expand on what

 

 12   you said, which is absolutely correct, there is a

 

 13   research team in a pharmaceutical

 

 14   industry-sponsored study.  Beyond that team, within

 

 15   the company itself, there is also the equivalent of

 

 16   a data monitoring board which usually consists of

 

 17   the project clinician, the safety officer and the

 

 18   statistician as a minimum.  The data is looked at

 

 19   very frequently.  In addition to that, there are

 

 20   periodic presentations of the safety data to larger

 

 21   committees within the company and then there is an

 

 22   opportunity to also present that data, if there are

 

 23   any red flags, to the scientific advisory board of

 

 24   external advisors which usually meets three to four

 

 25   times a year depending on the situation.

 

                                                               185

 

  1             DR. SANTANA:  Donna?

 

  2             DR. PRZEPIORKA:  Actually, it sounds like

 

  3   industry has a separate oversight; the organized

 

  4   groups have a separate oversight; NCI-sponsored

 

  5   studies will have a separate oversight.  What we

 

  6   haven't discussed is individual

 

  7   investigator-initiated studies at single

 

  8   institutions.  I think under those circumstances it

 

  9   might not be too disruptive to say, you know, at

 

 10   some point see if there is somebody who can give

 

 11   you an outside reality check before you go on to

 

 12   the next level.  It may not require convening an

 

 13   entire board but just sending a member to the IRB

 

 14   or to whatever institutional data safety monitoring

 

 15   committee might be available.

 

 16             But, you know, having conducted Phase I

 

 17   studies, one can get lulled into, okay, I have five

 

 18   more patients lined up; let's go to the next level

 

 19   before I really have all the data collected on

 

 20   safety.  It may be just enough to actually improve

 

 21   patient safety at that one institution.

 

 22             DR. SANTANA:  Yes, I am glad you mentioned

 

 23   that.  We tried to address that at St. Jude.  As an

 

 24   academic institution, we tried to address that too

 

 25   with some of our own Phase I studies.  So, we

 

                                                               186

 

  1   operated very similarly to what the Phase I

 

  2   Consortium is doing, and that is that if it is an

 

  3   institutional Phase I study the research team meets

 

  4   frequently to review, as the study is being

 

  5   conducted, what the safety concerns are; what is

 

  6   going on with the next escalation, etc., etc.

 

  7             Then there are two separate groups that

 

  8   also look at that.  There is a separate Phase I/II

 

  9   planning group that we have that includes

 

 10   disciplines from solid tumors, leukemia,

 

 11   transplantation and biostatistics, all the basic

 

 12   science people and they are also supposed to meet

 

 13   every month but in reality they probably meet every

 

 14   six weeks and all the studies are also actually

 

 15   presented very briefly.  So, the whole group knows

 

 16   where each study is going and what is happening

 

 17   with toxicity; what is happening with issues of

 

 18   accrual.  That is not truly separate because it is

 

 19   constituted by individuals from the same

 

 20   institution.

 

 21             The third layer is that even for Phase I

 

 22   studies-- if you saw in my flow diagram where data

 

 23   went, all the adverse events, independent of any

 

 24   type of study, also get reviewed by the clinical

 

 25   protocol scientific review group subcommittee which

 

                                                               187

 

  1   does not include any of the Phase I PIs.  They also

 

  2   make a judgment in terms of how that study is

 

  3   going; in terms of dose escalations; in terms of

 

  4   safety.  So, it is very similar and kind of a

 

  5   little bit of recapitulation of what the

 

  6   cooperative group is doing in terms of having other

 

  7   people look at it.  It is not totally independent

 

  8   in the sense that there is an outside group that

 

  9   looks at it.

 

 10             Having said that, also in some Phase I

 

 11   studies, like the gene transfer studies--when we

 

 12   get to the question of DSMB committees I was going

 

 13   to mention that, we have a definition of what gets

 

 14   referred to DSMB and one of the definitions is if

 

 15   there is a Phase I study that includes  gene

 

 16   transfer or a biologic that is potentially

 

 17   problematic, that will go to the DSMB although it

 

 18   is a Phase I study.  Barry and then Susan?

 

 19             DR. ANDERSON:  Also, being part of this

 

 20   review board at the cancer center data safety

 

 21   monitoring plan, anybody who is receiving a grant

 

 22   that might involve a clinical study as an

 

 23   individual also has to provide a data monitoring

 

 24   plan in order to receive the money for the grant.

 

 25             DR. SANTANA:  Susan?

 

                                                               188

 

  1             DR. WEINER:  Just a point of

 

  2   clarification, just to make sure that the following

 

  3   case is covered for Phase I and perhaps for Phase

 

  4   II in pediatrics, let's say a network of

 

  5   institutions that are doing combination therapy

 

  6   trials, pharmaceutical trials, and they are not

 

  7   being supported by NIH--presumably the institutions

 

  8   have assurances, etc., but the monitoring of that

 

  9   particular kind of trial.

 

 10             DR. SANTANA:  Do you want to address that

 

 11   because it is primarily coming from the issue of

 

 12   industry-sponsored small trials within two or three

 

 13   institutions?  Am I correct, Susan?

 

 14             DR. WEINER:  Or more.

 

 15             DR. SANTANA:  Or more.  Do you want to

 

 16   address that?

 

 17             DR. GRILLO-LOPEZ:  From the safety point

 

 18   of view, they are monitored in exactly the same way

 

 19   that I mentioned earlier.

 

 20             DR. WEINER:  Well, just in terms of the

 

 21   external terms.  So, the company sets up some

 

 22   external monitoring to review safety concerns--I

 

 23   mean, if it is two drugs--

 

 24             DR. GRILLO-LOPEZ:  Well, if it is a Phase

 

 25   I or Phase II trial usually there is no external

 

                                                               189

 

  1   review, external to the company review, other than

 

  2   that the company has to report to the FDA.  So,

 

  3   that is an external third party.  Also, the company

 

  4   has the possibility of presenting the safety

 

  5   information to the scientific advisory board which

 

  6   is also an external review board.

 

  7             If there is a Phase III randomized study,

 

  8   particularly a blinded study, most companies are

 

  9   opting to have an external independent data safety

 

 10   monitoring board following that study, or if it is

 

 11   a Phase II trial that is already randomized and

 

 12   blinded.

 

 13             DR. SANTANA:  Any other comments or

 

 14   questions?  Then we will move on to the next

 

 15   question which is asking us for advice on what

 

 16   circumstances would benefit from a data monitoring

 

 17   committee/data safety review board oversight?

 

 18             To try to address that, I think Barry had

 

 19   in one of his slides what some of the

 

 20   recommendations are from NCI regarding--or was it

 

 21   COG?  I don't remember that.

 

 22             DR. ANDERSON:  Recommendations from NIH.

 

 23             DR. SANTANA:  Do you want to expand on

 

 24   those, Barry?

 

 25             DR. ANDERSON:  In pediatrics the default

 

                                                               190

 

  1   seems to have some sort of monitoring committee, a

 

  2   more formalized monitoring committee because the

 

  3   recommendations were if they were complex--and if

 

  4   you have ever looked at an ALL study or anything

 

  5   else, they are pretty complex, and every study

 

  6   basically, if it is multi-institutional, which

 

  7   pediatrics for the most part usually are--if it is

 

  8   a vulnerable patient population, and we have our

 

  9   own separate part of the regulations just because

 

 10   pediatrics is a vulnerable patient population, and

 

 11   high-risk treatments--you know, a lot of the

 

 12   treatments that we use with stem cell transplants,

 

 13   etc., etc., are high risk.  So, because of all

 

 14   those issues coming up in a lot of cases, a data

 

 15   monitoring committee is involved.  It may be

 

 16   different than a DSMB that you were talking about

 

 17   for a Phase III randomized study because some of

 

 18   these monitoring committees also work for

 

 19   single-arm studies that may have early stopping

 

 20   rules that need to be interpreted, and that sort of

 

 21   thing as well.

 

 22             DR. SANTANA:  I would add two additional

 

 23   items to the list that Barry proposed.  As an

 

 24   institution, there are two other types of studies

 

 25   that we would refer for an independent data safety

 

                                                               191

 

  1   monitoring board.  One is any study that involves

 

  2   any type of gene transfer or biologic that

 

  3   potentially could present a hazard to children in

 

  4   the future.  Then, the second is a very unique type

 

  5   of study which is what we call the window study

 

  6   where an experimental therapy is given prior to

 

  7   conventional therapy and there is a limitation of

 

  8   time in which you can really do that to provide

 

  9   safety for the patients.  So, those kind of studies

 

 10   we would also refer to DSMB to provide oversight.

 

 11             Any other comments or questions on that?

 

 12   Yes?

 

 13             DR. GRILLO-LOPEZ:  A clarification, when

 

 14   you made your presentation you mentioned the makeup

 

 15   of your data monitoring board and you said it was

 

 16   the staff involved in the study itself, the

 

 17   principal investigator and perhaps some others

 

 18   around the principal investigator, and then some

 

 19   additional members outside of that group.  But

 

 20   should I interpret "outside" as within the

 

 21   cooperative group or completely external to the

 

 22   cooperative group?

 

 23             DR. ANDERSON:  It depends on whether you

 

 24   are talking about a DSMB or a DMC.  I mean, there

 

 25   has been some distinction there.  The DSMBs would

 

                                                               192

 

  1   be probably reflective of what industry uses when

 

  2   they have an outside independent one.  For the COG

 

  3   DSMBs there is a member or maybe two members of COG

 

  4   that are part of that but there are statisticians

 

  5   from other adult cooperative groups.  There are

 

  6   outside lay people that are part of it, and there

 

  7   is a government representative there.  It is set up

 

  8   such that the vote could never be carried by COG

 

  9   members.  And someone who would be perhaps a study

 

 10   investigator for a particular Phase III study, they

 

 11   would not be involved in discussions of their study

 

 12   if they happened to be also a COG representative to

 

 13   the group.

 

 14             For other data monitoring committees--I

 

 15   can't speak for Peter's group but for the NANT that

 

 16   data monitoring committee has one representative

 

 17   from that group or institutions that are conducting

 

 18   these early phase studies.  A number of people are

 

 19   COG members but they don't participate in these

 

 20   studies.  There are other people who are retired

 

 21   pediatric oncologists.  We have statisticians from

 

 22   outside the group.  We have lay people from outside

 

 23   the group.  So, again, we are not looking at Phase

 

 24   IIIs, we are looking at early studies.  Again, the

 

 25   predominant role is that you are outside of the

 

                                                               193

 

  1   people who are doing the investigations.  That,

 

  2   again, is for the interval of about every six

 

  3   months of formal review but also being there as a

 

  4   resource ongoing.

 

  5             The reviews that go on in the NANT group

 

  6   sort of on a more frequent basis could involve

 

  7   study investigators but it is usually the bigger

 

  8   group of other investigators that are part of the

 

  9   group but not responsible for that particular

 

 10   study.  So, there is some oversight in the sense

 

 11   that it is within the group but it is not the

 

 12   person who has the most vested interest that that

 

 13   single study succeed in one way or another.

 

 14             DR. GRILLO-LOPEZ:  It is probably

 

 15   worthwhile to mention that in industry today most

 

 16   Phase I and II studies have an enrollment period

 

 17   that ranges from 6 months to 12 months and perhaps

 

 18   not more than that.  So, the value of an external

 

 19   data safety monitoring board is limited because of

 

 20   your ability to actually give them trend

 

 21   information and so on when you have actually

 

 22   completed enrollment on the study.

 

 23             DR. PAZDUR:  I would just like to mention

 

 24   that we have a draft guidance on data safety

 

 25   monitoring.

 

                                                               194

 

  1             DR. SANTANA:  I think with that we will go

 

  2   to the last question, which is an open-ended

 

  3   question, are there additional recommendations for

 

  4   safety monitoring?  Peter?

 

  5             DR. ADAMSON:  I think the only one that

 

  6   came up earlier is that institutions don't have

 

  7   adequate resources to do this job well.  That is

 

  8   not unique to pediatrics but every layer of

 

  9   monitoring that gets put on an institution and

 

 10   investigator--you have to look if the resources are

 

 11   there to truly meet it.  I think in most

 

 12   institutions the resources are inadequate right

 

 13   now.

 

 14             DR. SANTANA:  I would echo that.  I think

 

 15   we started this morning's session with a comment

 

 16   about stewardship and I think stewardship includes

 

 17   financial resources so I think the regulatory

 

 18   agencies need to be very cognizant that if we are

 

 19   going to do this, there has to be a mechanism to

 

 20   provide monies to do this well.  There can't be

 

 21   mandates without monies to actually carry this out

 

 22   well.  Susan?

 

 23             DR. WEINER:  I have one additional

 

 24   comment, and that is that I think that the term

 

 25   "lay member" is fine but it seems to me that when

 

                                                               195

 

  1   one is reviewing pediatric trials there really

 

  2   ought to be a family member who is that lay person

 

  3   to help assess the safety of the situation.

 

  4             DR. SANTANA:  Good point.  Any other

 

  5   comments?  Any other guidance that the FDA wishes

 

  6   from us on this session?  If not, we are adjourned

 

  7   for the morning.  Thank you.  We will try to

 

  8   reconvene at about 1:15.

 

  9             [Whereupon, at 12:25 p.m., the proceedings

 

 10   were recessed for lunch, to reconvene at 1:20 p.m.]

 

                                                               196

 

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

 

  2             DR. SANTANA:  Let's go ahead and get

 

  3   started with the afternoon session in which we are

 

  4   going to talk about preclinical models and other

 

  5   data that we could extrapolate in terms of helping

 

  6   us design clinical studies.  Before we get started

 

  7   with the actual presentations, we need to go around

 

  8   the table again and re-introduce ourselves because

 

  9   there are new individuals who have joined the group

 

 10   and, hopefully, not many others have left.  So, can

 

 11   we start with Dr. Anderson, please?

 

 12             DR. ANDERSON:  Barry Anderson, from NCI

 

 13   CTEP.

 

 14             DR. HOUGHTON:  Peter Houghton, from St.

 

 15   Jude Research Hospital.

 

 16             DR. ADAMSON:  Peter Adamson, The

 

 17   Children's Hospital of Philadelphia.

 

 18             DR. HELMAN:  Lee Helman, Pediatric

 

 19   Oncology Branch, National Cancer Institute.

 

 20             DR. SMITH:  Malcolm Smith, Cancer Therapy

 

 21   Evaluation Program, NCI.

 

 22             DR. GRILLO-LOPEZ:  Antonio Grillo-Lopez,

 

 23   Neoplastic and Autoimmune Diseases Research

 

 24   Institute.

 

 25             MS. HAYLOCK:  Pam Haylock, oncology nurse

 

                                                               197

 

  1   and ODAC consumer representative.

 

  2             DR. PRZEPIORKA:  Donna Przepiorka,

 

  3   University of Tennessee, Memphis.

 

  4             MS. CLIFFORD:  Johanna Clifford, executive

 

  5   secretary for this meeting.  I am just curious, is

 

  6   Eric Kodish still on the line?  No.

 

  7             DR. SANTANA:  Victor Santana, pediatric

 

  8   oncologist at St. Jude's Children's Research

 

  9   Hospital, in Memphis, Tennessee.

 

 10             DR. REYNOLDS:  Patrick Reynolds,

 

 11   Children's Hospital of Los Angeles.

 

 12             MS. ETTINGER:  Alice Ettinger, nurse

 

 13   practitioner at St. Peter's University Hospital in

 

 14   New Jersey.

 

 15             DR. WILLIAMS:  Grant Williams, Oncology

 

 16   Drugs.

 

 17             DR. KEEGAN:  Pat Keegan, Oncology

 

 18   Biologics.

 

 19             DR. HIRSCHFELD:  Steven Hirschfeld, FDA.

 

 20             DR. DINNDORF:  Pat Dinndorf, Oncology

 

 21   Biologics.

 

 22             DR. DAGHER:  Ramzi Dagher, Division of

 

 23   Oncology Drug Products, FDA.

 

 24             DR. SANTANA:  Thank you.  With that, we

 

 25   will go ahead and get started with the first

 

                                                               198

 

  1   presentation, Dr. Paul Meltzer.

 

  2          What are Microarrays and How Can They Help Us

 

  3           with Clinical Studies in Pediatric Oncology

 

  4             DR. MELTZER:  What I am going to do is to

 

  5   very quickly give the members of the committee a

 

  6   tour of some of the clinically relevant

 

  7   applications of genomic technologies involving

 

  8   microarrays which may have a bearing on some of the

 

  9   issues that you are considering today.  I will do

 

 10   that in the fashion of a very brief overview of

 

 11   technology in a few specific examples, and give you

 

 12   my impression of some of the issues that would have

 

 13   to be overcome for this information to be evaluable

 

 14   in clinical trials.

 

 15             Array technologies have now been around

 

 16   for several years, and the ones that I am going to

 

 17   talk about mainly today are actually becoming

 

 18   rather mature, and it is now possible to generate

 

 19   data with these technologies which can be

 

 20   considered sort of archival quality that will serve

 

 21   as a long-term source of information about the

 

 22   diseases that are being looked at.

 

 23             There is some the excitement around these

 

 24   technologies, as indicated by this slide which just

 

 25   shows the number of citations in PubMed on

 

                                                               199

 

  1   microarrays from the inception of the modern

 

  2   technology for microarray expression profiling

 

  3   through last years.  There has been an exponential

 

  4   growth in the number of publications that cut

 

  5   across all areas of biomedical research.  There has

 

  6   been a tremendous amount of interest and activity

 

  7   in data generation, importantly, for you to

 

  8   consider.

 

  9             The reason that this momentum has built up

 

 10   has been based on the availability of the human

 

 11   genome sequence which now allows a whole genome

 

 12   approach to identifying the genes expressed in

 

 13   tumor tissue samples or in the context of other

 

 14   types of biological samples.  Of course, this will

 

 15   include drug targets and, indeed, it should include

 

 16   every conceivable protein drug target, as well as

 

 17   gene expression signatures which represent a

 

 18   cellular readout that is associated with important

 

 19   clinical or biological properties of cancers.  I

 

 20   will try to explain this concept with just a few

 

 21   examples in a moment.

 

 22             There are a number of different microarray

 

 23   technologies and I am just going to be touching on

 

 24   the two that are underlined because these are the

 

 25   ones that are in most widespread use today really

 

                                                               200

 

  1   throughout the world.  At the top of the list, and

 

  2   mainly what I will be talking about, is expression

 

  3   profiling, measuring the expression of large

 

  4   numbers of genes in parallel in a given biological

 

  5   sample.

 

  6             It is important to note that there are

 

  7   other array technologies coming along which are

 

  8   likely to have a role of some type in clinical

 

  9   application, and that includes microarrays to

 

 10   determine DNA copy number in tumors, or CGH arrays,

 

 11   microarrays which can determine DNA polymorphisms,

 

 12   commonly referred to as SNP chips.  I am going to

 

 13   touch briefly on tissue microarrays because they

 

 14   have emerged as a very important confirmatory

 

 15   mechanism for the RNA-based expression arrays which

 

 16   are also potentially of clinical importance.  Of

 

 17   course, protein arrays, various forms of

 

 18   proteomics, are important and I am not going to

 

 19   talk about that.

 

 20             It is important for you to realize that

 

 21   there is a tremendous amount of gene expression

 

 22   data, mainly from adults, which has already been

 

 23   generated with these technologies, and a great deal

 

 24   of this is already publicly available in databases

 

 25   that are universally accessible.

 

                                                               201

 

  1             So, this is just what one form of

 

  2   microarray looks like, basically a glass microscope

 

  3   slide on which DNA probes have been deposited.  I

 

  4   won't dwell on the technology, other than to point

 

  5   out the important feature, and there are several

 

  6   different embodiments of the technology but the

 

  7   important feature is that we now can look at the

 

  8   entire human genome, or animal genome if you are

 

  9   talking about an animal model, cramming in the

 

 10   entire genome on a single microarray chip and it is

 

 11   possible to interrogate this chip, to use it to

 

 12   interrogate a biological sample to look at

 

 13   expression of all the different genes in the human

 

 14   genome in a biological specimen.  For those of you

 

 15   who are into gene expression, you know that there

 

 16   are subtleties involving, let's say, splice

 

 17   isoforms and, indeed, that is being looked at with

 

 18   this type of technology as well.  So, you can

 

 19   really get a very detailed picture of expression

 

 20   across the genome at the RNA level with this

 

 21   technology, and one that is actually remarkably

 

 22   accurate and carries with it quite a nice snapshot

 

 23   of an individual biological sample.

 

 24             So, what are some of the potential

 

 25   connections between this information and cancer

 

                                                               202

 

  1   therapeutics?  The first I would mention is to

 

  2   increase the precision in tumor diagnosis to

 

  3   complement additional pathologic techniques, and

 

  4   perhaps to identify and define subsets that haven't

 

  5   been previously recognized in previously thought to

 

  6   be homogeneous tumor groups; to measure the

 

  7   expression of drug targets; to recognize

 

  8   signatures, and I will expand on this in a minute,

 

  9   which might be associated with the activity of a

 

 10   particular drug target; to identify features in the

 

 11   gene expression profile which might be related to

 

 12   drug sensitivity or resistance; and potentially to

 

 13   monitor or predict toxicity.

 

 14             Now, there are subtleties and, in fact, it

 

 15   is actually an extremely complex topic, the

 

 16   analysis of microarray data that I am not really

 

 17   going to touch on, but it is important for me to

 

 18   point out to you that aside from simply scoring in

 

 19   a simple kind of plus/minus way for the presence of

 

 20   a given gene or target, all of these types of

 

 21   analyses require a training set of tumors to

 

 22   identify the relevant genes and to develop a

 

 23   scoring algorithm which can be used to look at

 

 24   these various types of readouts.

 

 25             Another very important feature of this

 

                                                               203

 

  1   data is that if you have full genome data it is

 

  2   comprehensive.  It is intrinsically comprehensive.

 

  3   There are only so many genes in the human genome;

 

  4   there won't be more in five years than there are

 

  5   now, or in 10 years or in 20 years.  That is why

 

  6   the data has a very nice archival quality to it.

 

  7   So, it can be reanalyzed in the future with respect

 

  8   to novel targets or signatures that might be

 

  9   identified so you basically have data that really

 

 10   won't go stale so long as it is collected in a

 

 11   state-of-the-art fashion and is appropriately

 

 12   archived.

 

 13             This slide just outlines the strategy that

 

 14   is used in microarray studies.  You start with the

 

 15   whole genome and look at a very large number of

 

 16   genes, so tens of thousands of genes across many

 

 17   samples to develop profiles that occur in a

 

 18   particular clinical situation.  Then you go through

 

 19   some process of gene selection to identify those

 

 20   genes which separate tumors or patients into groups

 

 21   according to the particular question that is being

 

 22   asked, whether it be drug response, toxicity or a

 

 23   diagnostic question, genes that are associated with

 

 24   a particular target activity, and so on.

 

 25             You then have to go through a process of

 

                                                               204

 

  1   validation, frequently involving a new sample set

 

  2   and reiterating this process to validate it, and

 

  3   also probably validating it with other technologies

 

  4   such as RT PCR, quantitative PCR or

 

  5   immunohistochemistry or RNA in situ hybridization

 

  6   or something like that to validate the results.

 

  7   You might want to proceed to a clinical assay

 

  8   development, and it is very important to point out

 

  9   that much of the momentum in the development of

 

 10   clinical assays based on this type of information

 

 11   involves not microarrays but other forms of

 

 12   multiplex gene analysis which might involve, for

 

 13   example, a PCR-based method.

 

 14             So, this is the overall approach and here

 

 15   are going to be a couple of very quick examples.

 

 16   This is from a study we published several years ago

 

 17   identifying groups of genes that separate for

 

 18   common pediatric cancers, Ewing's sarcoma and

 

 19   neuroblastoma, rhabdomyosarcoma and Burkitt's

 

 20   sarcoma.  Color-coded here and at the top of this

 

 21   clustergram, each of these little groups of red

 

 22   squares represents groups of genes that separate

 

 23   these groups of tumors and can be used to diagnose

 

 24   them with a high degree of accuracy.

 

 25             The important point about this slide is

 

                                                               205

 

  1   that out of a large number of thousands of genes,

 

  2   the genes that were necessary to give a perfectly

 

  3   accurate call involved a very small number of

 

  4   genes, about a 100 genes, 96 to be precise, which

 

  5   were identified by a process of gene minimization.

 

  6   So, that is the bottom line of everything that you

 

  7   will see in the literature or hear about, that one

 

  8   doesn't need tens of thousands of genes to answer a

 

  9   question.  If it is possible to answer it, usually

 

 10   a very small number of genes, less than 100, will

 

 11   be sufficient to accomplish what you want and

 

 12   sometimes as few as two.

 

 13             I am going to give two quick examples that

 

 14   illustrate these features in detecting therapeutic

 

 15   targets by microarrays, one in gastrointestinal

 

 16   stromal tumor, or GIST, and the other is breast

 

 17   cancer which involved a couple of studies that were

 

 18   from our lab.

 

 19             In the case of GIST, here were are seeing

 

 20   the separation of gastrointestinal stromal tumor

 

 21   from non-GIST sarcomas with, again, the minimal

 

 22   number of genes necessary to establish the

 

 23   separation.  The important point for today's

 

 24   discussion is that when we looked at the top genes

 

 25   we found that the KiT oncogene was actually the

 

                                                               206

 

  1   number one gene.  So, we both could score the

 

  2   presence and assess its relative importance in

 

  3   characterizing this particular tumor in one

 

  4   process, and one can do this in respect to any

 

  5   property of a tumor that you choose.  So, this is

 

  6   an example of scoring a single gene out of

 

  7   microarray data.

 

  8             If you will forgive me for introducing an

 

  9   adult example, I will now give you an example that

 

 10   indicates how you might work--

 

 11             Oh, this is just to show how KiT looks on

 

 12   a heat map of GIST versus non-GIST.  You see this

 

 13   very uniform pattern of KiT expression.

 

 14             I will give an example now of how you

 

 15   would look at a signature for gene expression using

 

 16   the estrogen receptor in breast cancer which, of

 

 17   course, is a very nice molecular target widely used

 

 18   in breast cancer therapy.  The point here is that

 

 19   there is a distinct pattern of gene expression in

 

 20   breast cancer that separates the positive from

 

 21   negative tumors very sharply, and everybody who

 

 22   looks at these tumors has found exactly the same

 

 23   result.  It is the strongest feature in gene

 

 24   expression profile of breast cancer.

 

 25             Importantly, it is possible to actually

 

                                                               207

 

  1   predict the value of the protein measurement for ER

 

  2   in a tumor specimen from the gene expression

 

  3   profile using a number of genes to make that

 

  4   prediction excluding the estrogen receptor itself.

 

  5   So, you can actually plot on this figure the actual

 

  6   ER level in the little magenta circles, and the

 

  7   predicted value based on the gene expression

 

  8   profile based on a group of several hundred genes

 

  9   in these tumors.  So, there is a signature that

 

 10   goes with the presence and function of this

 

 11   particular drug target that can be read out using

 

 12   multiple genes.  Similar observations have been

 

 13   made for other targets.  So, this is an example of

 

 14   a multiple gene predictor.

 

 15             The bottom line here is that microarrays

 

 16   can measure therapeutically relevant genes either

 

 17   as individual genes or as complex signatures, and

 

 18   expression profiling then can reveal both the

 

 19   presence of a target and measure relative abundance

 

 20   within the cell at the RNA level.  Finally, a

 

 21   signature related to target function can reveal its

 

 22   level of biological activity, as in the ER example.

 

 23             I just want to take a couple of moments to

 

 24   talk about tissue microarrays because I think these

 

 25   are very important and very accessible from a

 

                                                               208

 

  1   technological point of view.  A tissue mircoarray

 

  2   is simply an array taken from paraffin blocks from

 

  3   patient samples, assembled into an array which can

 

  4   then be sliced to produce many slides that can be

 

  5   assayed for various markers.  The power of this

 

  6   technology is that, in contrast to the DNA

 

  7   microarray in which we measure thousands of genes,

 

  8   for each tissue specimen in the tissue microarray

 

  9   we can measure one gene in thousands of specimens

 

 10   very rapidly.  So, these are very powerful tools

 

 11   for the validation of findings for genomic surveys

 

 12   and potentially for translating them into clinical

 

 13   studies.

 

 14             Just to emphasize the tremendous advantage

 

 15   that we gain from using these arrays, it arises

 

 16   from taking a large number of paraffin blocks and

 

 17   condensing them down into one very affordable,

 

 18   economical package where we can survey single

 

 19   tumors with a slice from any individual tissue

 

 20   microarray.  So, it is a very powerful technology

 

 21   that I think can be quite useful.

 

 22             So, how might these technologies be

 

 23   implemented in clinical trial designs?  I just want

 

 24   to take a moment to give you some perspective.

 

 25   First of all, to reiterate, detection of individual

 

                                                               209

 

  1   targets is really simple.  That is not difficult

 

  2   and is very straightforward and should pose no real

 

  3   challenge.  However, in terms of using this for

 

  4   pediatric cancers, we have a problem in that so far

 

  5   only limited data is available on pediatric cancers

 

  6   in the public repositories and that would be one of

 

  7   the major obstacles.  Indeed, very minimal data

 

  8   exists relative to any question of toxicity, and

 

  9   these are issues that are just beginning to be

 

 10   seriously looked at in adults and, to my knowledge,

 

 11   haven't been examined in children at all.  As far

 

 12   as I can see, implementing tissue collection

 

 13   protocols and microarray analysis as part of

 

 14   ongoing trials would be a necessity to overcome

 

 15   this limitation.

 

 16             Tumor tissue sampling is essential to get

 

 17   a picture of the tumor but I am not sure that it is

 

 18   necessary to have serial sampling.  It would be in

 

 19   principle nice to know what happens in the residual

 

 20   tumors of patients who don't respond to therapy but

 

 21   in principle this should be predictable from the

 

 22   initial signature.

 

 23             It is also interesting to speculate that

 

 24   useful information regarding toxicity may

 

 25   potentially be obtained from blood samples for

 

                                                               210

 

  1   example, but the data to support this concept is

 

  2   extremely limited at the present time.

 

  3             Finally, again to reiterate, complex

 

  4   questions such as the prediction of response or

 

  5   toxicity require a training set and can't be

 

  6   answered a priori or predicted from a bunch of

 

  7   array data.  So, if we want to talk about taking

 

  8   array data from an archive and predicting what

 

  9   might happen in those patients in response to a

 

 10   particular agent, we really don't have a way to do

 

 11   that at the present time.  The only way we can

 

 12   really examine that is to have samples annotated

 

 13   with respect to that clinical question.  So, that

 

 14   is basically what I had to say.  Thank you.

 

 15             DR. SANTANA:  Thank you.  We will have

 

 16   some opportunity during the discussion period to

 

 17   address some questions.  I think Dr. Peter Adamson

 

 18   is next.  Peter?

 

 19        Advantages and Limitations of Cell Culture Models

 

 20                  in Pediatric Drug Developments

 

 21             DR. ADAMSON:  For those of you who

 

 22   remember Monty Python and now for something

 

 23   completely different, whereas microarrays are

 

 24   approaching their tenth birthday, cell culture

 

 25   models are probably approaching retirement age. 

 

                                                               211

 

  1   So, what I thought I would do is speak briefly

 

  2   about some of the advantages and limitations of

 

  3   these models.  Historically, they have been

 

  4   controversial as well as helpful.  I think many of

 

  5   the issues that occurred historically are still

 

  6   issues today.

 

  7             To really understand that, I want to take

 

  8   you through a very brief history of cell culture

 

  9   models in the context of drug development.  In

 

 10   looking back, probably the clonogenic assay is a

 

 11   good starting point as far as how these models have

 

 12   been used.  This was work done by Hamburger and

 

 13   Salmon, published back in 1977 in Science.  What

 

 14   they were able to show was that they could take a

 

 15   number of primary human tumors and grow them up in

 

 16   a cell culture matrix.

 

 17             This is a photo micrograph from their

 

 18   publication.  Definition tumors have different

 

 19   colony formations but the concept was that these

 

 20   represented tumor stem cells, and stem cells were

 

 21   the renewal source and they served as a seat of

 

 22   metastatic spread, and cytotoxicity in this assay

 

 23   was going to be proportional to cytotoxicity in

 

 24   vivo.  If you didn't get at the stem cell, you

 

 25   weren't going to have an effective anti-cancer

 

                                                               212

 

  1   treatment.

 

  2             The way the clonogenic assay worked when

 

  3   it came to cytotoxicity is you would expose your

 

  4   culture media to various concentrations of drugs

 

  5   and then look at the effect on colony formation,

 

  6   look at the clonogenic assay.

 

  7             Predating the clonogenic assay there were

 

  8   other mechanisms to try to look at cell growth and

 

  9   behavior in vitro.  The tritiated thymidine assay

 

 10   was probably the most common one.  This was a

 

 11   pretty straightforward approach where you would

 

 12   tritiate thymidine and measure the incorporation

 

 13   into dividing cells.  It basically was a

 

 14   measurement of S-phase cells and it quantified that

 

 15   simply by counts per minute with a radioactive

 

 16   label.

 

 17             There were clearly limitations really to

 

 18   both of these approaches.  The clonogenic assay was

 

 19   very labor intensive and there were a number of

 

 20   investigators who, despite that hurdle, ran an

 

 21   incredible number of assays looking for activity of

 

 22   cytotoxic agents.  But the reality was that it was

 

 23   really not readily amenable to high throughput.

 

 24             Conversely, the tritiated thymidine,

 

 25   although there were the limitations of just using

 

                                                               213

 

  1   the radioactive label, was also a non-clonogenic

 

  2   method.  You are looking really at a different

 

  3   endpoint.

 

  4             Then the field began to change and began

 

  5   to change based on a paper by Mossman, an

 

  6   immunologist, in The Journal of Immunologic

 

  7   Methodology, in 1983 when he described what is an

 

  8   assay familiar to almost everyone, the MTT assay

 

  9   which was a colorimetric assay for cellular growth

 

 10   and survival.  In this assay a salt, MTT, when

 

 11   incubated with viable cells in the mitochondria

 

 12   undergoes a ring opening and produces a purple

 

 13   salt, formazan.  Then you solubilize this; you get

 

 14   a purple color and you put this in a plate reader

 

 15   and the intensity of the optical density is

 

 16   proportional to the cell number.  This assay really

 

 17   began to change a lot of what was happening in the

 

 18   world of cell culture and cytotoxicity.

 

 19             Perhaps in part where it had a great

 

 20   impact was at the NCI which, at this time, was

 

 21   looking at moving from their historic way to screen

 

 22   compounds for anti-cancer activity to what became

 

 23   known as the NCI 60 cell line screen.  This is a

 

 24   typical output on a plot of logarithmic

 

 25   concentrations of a drug as well as survival.  As

 

                                                               214

 

  1   many people have noted in the past, the 60 cell

 

  2   line incorporated a number of

 

  3   malignancies--leukemia, non-small cell, small cell

 

  4   and so forth, but there was ne'er a pediatric

 

  5   malignancy on this list.  There were many efforts

 

  6   made to try and change that and probably, in

 

  7   hindsight, it was probably just as well that we

 

  8   didn't.

 

  9             Nonetheless, in the late '80s, early '90s

 

 10   and even today there are a large number of

 

 11   clonogenic assays that were based on the MTT, XTT.

 

 12   The SRB assay, sulforhodamine blue, was the one

 

 13   that the NCI eventually adopted; historically

 

 14   trypan blue uptake in viable versus non-viable

 

 15   cells; and the list goes on and on.  Each of these

 

 16   has various advantages and various disadvantages

 

 17   but ultimately they are all measuring a very

 

 18   similar endpoint and these are non-clonogenic

 

 19   assays.

 

 20             At this point it is helpful to step back

 

 21   and say, well, what are non-clonogenic assays, when

 

 22   it comes to drug development, really telling us?

 

 23   What principles do they rest on?  Taking some

 

 24   liberties, I think these are the assumptions that

 

 25   are made.  As you can see, many of these

 

                                                               215

 

  1   assumptions are supported by data, others less so

 

  2   as we work down the list.  But the non-clonogenic

 

  3   assay is really a measurement of viable cell number

 

  4   and almost all the non-clonogenic assays do that to

 

  5   a reasonably good degree.

 

  6             Many of these have been correlated which

 

  7   is considered in vitro the gold standard, the

 

  8   clonogenic assay.  Again, not all of them, and it

 

  9   is very cell line dependent how well that

 

 10   correlates.  But then one starts making larger

 

 11   leaps.  That is, that the clonogenic assay somehow

 

 12   is correlated to in vivo cell growth and in vivo

 

 13   cell growth that is somehow correlated to the tumor

 

 14   growth in the patient.  So, when you start up here

 

 15   you have a long list to go down as far as what we

 

 16   are asking an assay to do as far as being able to

 

 17   predict or not predict what is going to happen in a

 

 18   patient.

 

 19             Let me talk about some of the potential

 

 20   uses.  I mentioned drug discovery and this is an

 

 21   output from a more recent NCI screen.  This has

 

 22   advanced as far as the type of information that

 

 23   comes back.  There is a compare algorithm that can

 

 24   talk about mechanism of action, and so forth, but

 

 25   if you put it in the broader context of drug

 

                                                               216

 

  1   discovery, this is not how drugs are discovered

 

  2   today.  I mean, in industry today you have a

 

  3   target; you develop an assay for a target and you,

 

  4   hopefully, have an assay that is amenable to high

 

  5   throughput.  For the most part, outside of the 60

 

  6   cell line screen, this is not how drugs are being

 

  7   discovered.

 

  8             But cell culture models are still useful

 

  9   in a number of areas.  You can study cellular

 

 10   pharmacology.  You can certainly study mechanism of

 

 11   action of drugs in these models, as well as

 

 12   evaluate drug resistance.

 

 13             Now, as pediatric tumor models, they have

 

 14   historically and continue to serve at some level as

 

 15   a screening for drug activity, but you can also ask

 

 16   dose or, more appropriately, concentration schedule

 

 17   dependent questions in cell culture models and one

 

 18   can evaluate drug combinations in these models.

 

 19             There are, not surprisingly, limitations.

 

 20   Some of these limitations are unique to in vitro

 

 21   models; some can be transferred over to in vivo

 

 22   models.  We know that cell lines undergo

 

 23   transformation to allow for in vitro growth.  For

 

 24   in vitro drugs that require metabolic activation or

 

 25   have active metabolites, you are likely to miss

 

                                                               217

 

  1   that.  You are not likely to be able to pick that

 

  2   up given the nature of the in vitro model.

 

  3             There are clearly potential differences in

 

  4   drug exposures in these in vitro models.  They can

 

  5   range from differences in protein binding.  Drug

 

  6   disposition is incredibly difficult to try to model

 

  7   in vitro.  You basically dump the drug in and you

 

  8   let it sit there for a period of time.  That is not

 

  9   what happens in a patient as far as how drug is

 

 10   cleared.  There are certainly differences in tumor

 

 11   micro-environment or lack of vascularization and

 

 12   hypoxia.  There are methods, and Pat has looked at

 

 13   some methods, to try to compensate for that in in

 

 14   vitro models to try to better reflect what is

 

 15   happening in vivo, and there are many other

 

 16   limitations.

 

 17             With that background, there are still some

 

 18   advantages to these models.  Relatively speaking,

 

 19   these are not labor intensive models.  They are

 

 20   relatively low cost and they are amenable to

 

 21   moderate throughput.  In addition, because of

 

 22   these, you have the ability to study multiple cell

 

 23   lines and I think, perhaps as we move forward in

 

 24   product oncology, the ability to study multiple

 

 25   combinations of drugs.

 

                                                               218

 

  1             One advantage of the in vitro model I

 

  2   think over other models is that it is probably the

 

  3   only model system that is mathematically amenable

 

  4   to defining synergy, additivity or antagonism.  It

 

  5   becomes very complex in other systems to really

 

  6   know if something is synergistic or not.  There are

 

  7   a number of accepted methods to do that in an in

 

  8   vitro system.

 

  9             So, let me start there and I am just going

 

 10   to share three very basic examples of in vitro

 

 11   models and what they can do, and I think I will be

 

 12   commended then for picking up the pace as far as

 

 13   getting us back on whatever time line we should

 

 14   have been on.

 

 15             The first one is determination of synergy.

 

 16   I know folks in the room know this, there is a

 

 17   problem with a simple addition method.  If your

 

 18   drug A kills 15 percent and drug B kills 25

 

 19   percent, well then, if the combination kills more

 

 20   than 50 percent it is synergistic.  Well, it

 

 21   doesn't take much to realize that you run into a

 

 22   problem pretty quickly if drug A kills 70 percent

 

 23   and drug B kills 70 percent.  You can't simply add

 

 24   them up.  We can't just say, aha, it is

 

 25   synergistic; it is more than the sum.  That is what

 

                                                               219

 

  1   we are sometimes left with, with in vivo models but

 

  2   it is very difficult to know that.  There are a

 

  3   number of mathematical approaches and these get

 

  4   debated constantly in journals that I don't like to

 

  5   read--

 

  6             [Laughter]

 

  7             --but they do get debated.  One of the

 

  8   more accepted models is the median effect model.

 

  9   There is now software that really can make this

 

 10   very user friendly and straightforward.  But if you

 

 11   have different drugs you first look for a rational

 

 12   effect as a concentration of dose and you do that

 

 13   with one drug; you lay on the other and you lay on

 

 14   the third, and then you realize you can't see what

 

 15   is going on.  So, you transform the data and you

 

 16   get what is called a median effect plot.  From the

 

 17   median effect you can calculate what is called a

 

 18   combination index.  Please don't try to figure this

 

 19   out from the graph, but let me tell you that the

 

 20   software will basically tell you, yes, it is

 

 21   synergistic or it is additive, or no, in fact, it

 

 22   is antagonistic.  There are other methods and

 

 23   probably all of them are reasonable methods to look

 

 24   for whether a combination is going to be

 

 25   synergistic.

 

                                                               220

 

  1             Other examples, and this is probably where

 

  2   this has been most widely used, that is, is this

 

  3   drug that is being developed in adult malignancies

 

  4   relevant to pediatric malignancies?  Does it have

 

  5   activity in pediatric tumors?

 

  6             So, I chose a relatively recent example

 

  7   that Beth Fox is working on at the NCI, epothilone

 

  8   B, a Bristol-Myers drug.  This is an analog that

 

  9   binds tubulin.  It stabilizes microtubules by

 

 10   inhibiting tubulin depolymerization, blocks mitosis

 

 11   and causes apoptosis.  Interestingly, this drug is

 

 12   cytotoxic in Taxane resistant tumors, as well as in

 

 13   cell lines that over-express MDR.  So there was an

 

 14   interest certainly in the pediatric community as

 

 15   far as is this a drug that we should be looking at.

 

 16             So, what one can do is one can look in

 

 17   vitro.  In general, it is always helpful to have

 

 18   some sort of reference base to compare your drug

 

 19   with.  In this case, we compared it to other

 

 20   microtubule toxins, paclitaxel, vincristine and

 

 21   vinorelbine and looked at the concentrations that

 

 22   were required to produce cytotoxicity in an in

 

 23   vitro model.  You can look at these and you can

 

 24   say, well, for these drugs, in fact, these are

 

 25   concentrations that fall within the range achieved

 

                                                               221

 

  1   in patients, and then you look at the drug in

 

  2   question and say, well, these are the

 

  3   concentrations that, if this model is predictive,

 

  4   one might anticipate needing as far as a relative

 

  5   effect and one can ask if there is adult Phase I

 

  6   data or are these relevant concentrations.

 

  7             In addition, one can do some

 

  8   pharmacodynamic work.  In this case, one can look

 

  9   at the concentrations that were effective.  Were

 

 10   you hitting your target in a very endpoint type of

 

 11   way before cytotoxicity?  What was the effect on

 

 12   the polymerization versus non-polymerization?  That

 

 13   is what Beth did in this study.  So, it is helpful

 

 14   as far as an inexpensive way to look across a panel

 

 15   of cell lines to get some idea that this drug may

 

 16   have some relevance.

 

 17             I think an area that we probably need to

 

 18   do more work on is integration with new agents.  I

 

 19   am going to choose leukemia as an example here.

 

 20   For those of you who don't do this on a regular

 

 21   basis, this, in one slide, is what childhood acute

 

 22   lymphoblastic leukemia therapy looks like with

 

 23   different phases of therapy from induction through

 

 24   consolidation, interim maintenance, all the way

 

 25   through maintenance to just over three years.

 

                                                               222

 

  1             As you can see, in each of these phases we

 

  2   treat children with anywhere from six to eight

 

  3   different cytotoxics.  Then, on this backbone of

 

  4   very successful therapy that is toxic and is not

 

  5   curing all children, along comes a new drug that

 

  6   has made its way through Phase I and Phase II and

 

  7   clearly has efficacy.  The question is, is it going

 

  8   to improve outcome?  The question is, aha, here is

 

  9   our new drug, and this drug in this case is the

 

 10   prodrug 506U, and now what?  And "the now what" is

 

 11   not an easy question to answer.  Where do you put

 

 12   it?  What are the risks and benefits of putting it

 

 13   in any one place?  We actually were confronting

 

 14   this problem, and still are with this drug as far

 

 15   as how do we integrate this into successful

 

 16   front-line therapy to ask a Phase III question?

 

 17             Well, we have the advantage that 506U is

 

 18   actually a drug that is a very old drug that has

 

 19   only clinically come to our attention in the last

 

 20   decade.  Work done by  Trudy Allen many, many years

 

 21   ago, beginning in the '50s and extending through

 

 22   the '60s taught us a whole--and a number of other

 

 23   investigators.  And, one thing that came to light

 

 24   with anti-metabolites was that there was a

 

 25   potential drug interaction, a negative interaction

 

                                                               223

 

  1   with asparaginase.  It turns out that for other

 

  2   drugs there is a very sequence-dependent drug

 

  3   interaction.  So, we asked ourselves, okay, we are

 

  4   using asparaginase at a number of points in this

 

  5   therapy, is that a potential problem?

 

  6             You can look in vitro and begin to get an

 

  7   answer to that.  So, in this set of experiments we

 

  8   did sequential exposure.  Nelarabine is 506U, so

 

  9   first exposing to nelarabine and then following

 

 10   with asparaginase, in this case, because this is in

 

 11   vitro and asparaginase is an enzyme, simply

 

 12   changing over to asparagine-deficient media and

 

 13   then asking the reverse sequence question at least

 

 14   in one cell line--and this is early work that is

 

 15   going to be presented at AACR in a couple of weeks,

 

 16   but in this case there is, indeed, a red flag.  If

 

 17   you expose cells to asparaginase before you expose

 

 18   them to 506U you are going to have as much as a one

 

 19   log decrease in effectiveness.  So, this is an

 

 20   important piece of information when it comes for us

 

 21   to try to determine what we should attempt to do

 

 22   and what we should avoid doing.  This is far from

 

 23   comprehensive and, again, there are only two cell

 

 24   lines and one cell line really didn't have a

 

 25   significant effect.  We have to do more work.  But,

 

                                                               224

 

  1   again, these models might help us in trying to

 

  2   understand how to integrate new agents on the

 

  3   backbone of effective therapy that we currently use

 

  4   in children.

 

  5             I want to just share a few perspectives in

 

  6   closing.  I will preface it by saying this was not

 

  7   a comprehensive talk on cell culture models and

 

  8   these are as much opinions as they are accepted

 

  9   fact.

 

 10             In vitro models are a cost efficient

 

 11   method to search for activity, but

 

 12   mechanistic-based approaches likely will have a

 

 13   higher yield.  In other words, drug discovery has

 

 14   moved on from screening I think in cell culture

 

 15   systems.

 

 16             In vitro  models can, however, further our

 

 17   understanding of drug action in pediatric tumors,

 

 18   and the moderate throughput is advantageous,

 

 19   especially when studying drug combinations.  I

 

 20   showed you that for leukemia we treat with eight or

 

 21   nine drugs.  It will become a nightmare trying to

 

 22   figure out all the combinations but with in vitro

 

 23   models you at least have a chance of grappling with

 

 24   some of the major issues there.

 

 25             For most cytotoxic agents, if it does not

 

                                                               225

 

  1   work in vitro it will not work in vivo.  So, the

 

  2   negative predictive value for most cytotoxics is

 

  3   pretty good.  If you can't kill the cell in the

 

  4   dish you probably shouldn't invest a lot of energy

 

  5   if this is a cytotoxic agent.

 

  6             Correlated to that, if it takes a

 

  7   super-pharmacologic concentration in vitro to have

 

  8   an effect, it will likely not fare well in vivo.

 

  9   For the most part, you can kill cell cultures with

 

 10   anything if you put enough in so you do have to put

 

 11   it in the context of are these concentrations

 

 12   relevant concentrations.

 

 13             Lastly, and this is where we probably fall

 

 14   down most often, if it works well in vitro there is

 

 15   a reasonable likelihood that it will do absolutely

 

 16   nothing in vivo.  That is true of a lot of models

 

 17   and it is certainly true of cell culture models.

 

 18   So, I will stop there and let the program continue.

 

 19             DR. SANTANA:  Thank you, Peter.  We have a

 

 20   few minutes for questions because we have to do two

 

 21   things, we have an open public hearing if anybody

 

 22   wants to speak and we also have to switch laptops.

 

 23   So, there is opportunity to address any questions

 

 24   to Dr. Adamson and Dr. Meltzer now.  I have a

 

 25   question for Dr. Meltzer, you kind of hinted at the

 

                                                               226

 

  1   end of your talk about an issue of peripheral blood

 

  2   and, I read in between the lines surrogate use of

 

  3   peripheral blood.  Can you expand on what you

 

  4   meant?  Did you mean that you would take the tumor

 

  5   diagnosis, establish a profile, and do it also with

 

  6   peripheral blood and diagnosis but then only

 

  7   monitor peripheral blood as your surrogate?  Please

 

  8   go the microphone.

 

  9             DR. MELTZER:  What I really meant was

 

 10   monitoring toxicity, and the example that I know of

 

 11   that has the most effort is in actually monitoring

 

 12   for radiation toxicity.  There are patients who are

 

 13   extremely sensitive to radiotherapy and have severe

 

 14   toxicity and there are some tantalizing preliminary

 

 15   data from Stanford that suggest that you can tell

 

 16   the hypersensitive patients by gene expression

 

 17   profiling of their peripheral blood.  That is an

 

 18   approach that, to my knowledge, has not been really

 

 19   applied to chemotherapy and there may be an

 

 20   opportunity to do that.  So, I was really

 

 21   speculating.

 

 22             DR. SANTANA:  Dr. Reynolds?

 

 23             DR. REYNOLDS:  Peter, I think there are a

 

 24   couple of comments I want to make about what you

 

 25   said about the predictive value of these.  One is

 

                                                               227

 

  1   that I think there was a very interesting panel

 

  2   discussion at the AACR ERTC meeting in Boston this

 

  3   year about the predictive value of models in

 

  4   general.  It wasn't just in vitro, it was talking

 

  5   about animal models.  The conclusion was that they

 

  6   were basically non-predictive and, you know, no one

 

  7   had any magic models.

 

  8             At the same time, when you look at the

 

  9   publication that is coming out of the NCI 60 cell

 

 10   line screen, what they are saying is that the one

 

 11   thing that was somewhat predictive is if they have

 

 12   activity in multiple different cell lines, then

 

 13   that tended to give you some predictive value.  So,

 

 14   more is better in that setting.

 

 15             The third is that there are some

 

 16   well-established principles that have been

 

 17   discussed in the literature and often ignored that

 

 18   say that if you really can get two logs worth of

 

 19   activity, whether it is in an animal model or in an

 

 20   in vitro model, that may be somewhat predictive.

 

 21   In other words, there is a two-log threshold, which

 

 22   you didn't address.  And, I think when you talk

 

 23   about IC50s we clearly are not talking about

 

 24   multi-log assays or in the MPT system.

 

 25             So, I guess what I am suggesting is that I

 

                                                               228

 

  1   think that one reason why the predictive value of

 

  2   some of these has been less than we would all like

 

  3   is that, first of all, I think the systems still

 

  4   aren't optimized and I think they need to be done

 

  5   in multi-log systems and, secondly, as you pointed

 

  6   out, a number of us are studying things like

 

  7   physiological hypoxia and the impact on this.

 

  8   Certainly, as you pointed out very astutely, there

 

  9   must be consideration of what the pharmacological

 

 10   parameters you are going to see in a patient are

 

 11   when you approach these.

 

 12             Third, I think what we really need is to

 

 13   be doing them in more cell lines, not just one, two

 

 14   or three but we need a lot of them.  Once we get

 

 15   the right panels of biologic reagents in these

 

 16   systems and the right systems we might see the

 

 17   predictive value go up, and I don't think that we

 

 18   should exclude that possibility when we consider

 

 19   these.

 

 20             DR. SANTANA:  Dr. Grillo?

 

 21             DR. GRILLO-LOPEZ:  Another comment that I

 

 22   would like to make is that many of these models

 

 23   have been developed for chemotherapeutic agents and

 

 24   when you are dealing with a biological they may

 

 25   have no applicability whatsoever.

 

                                                               229

 

  1             DR. SANTANA:  Any other comments?

 

  2             [No response]

 

  3             We have a few minutes for an open public

 

  4   hearing so if there is anybody in the audience who

 

  5   wishes to address the committee, could you please

 

  6   come forward to the podium and identify yourself

 

  7   and any potential conflicts of interest, and make

 

  8   your statement?

 

  9             Well, if nobody is going to take the

 

 10   opportunity, then we will invite Dr. Houghton to

 

 11   proceed with the next presentation.

 

 12        Human Cell-Animal Xenografts: The Current Status,

 

 13            Potential and Limits of Informing us About

 

 14                         Clinical Studies

 

 15             DR. HOUGHTON:  I would like to thank Steve

 

 16   for inviting me.  When we were given the mandate or

 

 17   the subject of this afternoon's session, it was

 

 18   actually a clarifying moment to think about what

 

 19   sort of preclinical data is required or is of any

 

 20   use.  I think there are two ways of looking at what

 

 21   sort of preclinical data can be of use.  That is,

 

 22   use for us in sort of designing clinical trials as

 

 23   opposed to perhaps the information that would be

 

 24   required for the FDA to make some sort of decisions

 

 25   regarding the potential use of an agent.

 

                                                               230

 

  1             So, I think we can look at early drug

 

  2   discovery largely within defined standardized

 

  3   environments, either in drug discovery groups

 

  4   within companies where you have set protocols and

 

  5   set criteria for establishing whether an entity has

 

  6   adequate activity to progress to the next stage, or

 

  7   in the NCI screening program where, again, there is

 

  8   a set of protocols that drive the criteria for

 

  9   advancement of the compound.  The problem is that

 

 10   pediatric cancers are represented in neither

 

 11   entity.  They are obviously not going to be a focus

 

 12   of the pharmaceutical industry and, as Peter

 

 13   alluded to, despite multiple attempts they were not

 

 14   included in the NCI screening program.

 

 15             The consequences of preclinical data using

 

 16   pediatric models is generated essentially in an

 

 17   uncontrolled or non-regulated environment where

 

 18   everyone uses their own pet models, their pet

 

 19   design of experiments and, in fact, their own

 

 20   criteria for assessing whether or not they regard

 

 21   something as being active.  So, such data derived

 

 22   from experimental systems that are not validated,

 

 23   using experimental designs that are, again, not

 

 24   validated and interpretation of those results lacks

 

 25   consistency and rigor.

 

                                                               231

 

  1             So, taking advantage of the approaches

 

  2   that have been taken in industry and the idea of

 

  3   developing a consistent, criteria-driven approach a

 

  4   group, many of whom are represented here, under the

 

  5   leadership of Malcolm Smith, Barry Anderson and

 

  6   Peter Adamson, met during 2002 to consider what

 

  7   sort of screening program would be useful to

 

  8   implement that would allow us to identify drugs

 

  9   that are in the early clinical or just at the late

 

 10   preclinical stages from industry that might be

 

 11   useful in identifying drugs that would have

 

 12   specific application and perhaps should be

 

 13   prioritized for pediatric clinical testing.

 

 14             The schema is shown here and I am not

 

 15   going to go into detail on this because Malcolm is

 

 16   going to deal with this in somewhat more detail in

 

 17   his talk.  But the idea is to set up a panel of

 

 18   models so tumor A may be medulloblastomas and tumor

 

 19   B may be neuroblastomas, a panel of six to ten of

 

 20   these comprising either xenografts or heterografts

 

 21   of human cancers in immune incompetent mice, or

 

 22   where there are transgenic models to implement

 

 23   those within the screening program.  But the idea

 

 24   would be that we would have a framework where we

 

 25   can set the criteria for experimental design, set

 

                                                               232

 

  1   the criteria for assessing responses that may be

 

  2   more consistent and may generate data that would be

 

  3   of use not only to us as a group that are

 

  4   interested in developing clinical trials, but

 

  5   perhaps more appropriate use to a federal agency

 

  6   such as the FDA if they wanted to use such

 

  7   preclinical data.

 

  8             So, we were asked to look at the following

 

  9   categories of nonclinical data, and I am going to

 

 10   concentrate on pharmacology and pharmacokinetics

 

 11   efficacy and the aspect of using the models to

 

 12   identify pharmacodynamic endpoints that may be more

 

 13   amenable to analysis within the model systems than

 

 14   they are, certainly, in patients with solid tumors.

 

 15   The other aspect is to ask where such data fits in

 

 16   terms of development of drugs in the pediatric

 

 17   cancer realm.

 

 18             I think the models can be useful in

 

 19   identifying active agents and perhaps better

 

 20   analogs to optimize the administration schedules,

 

 21   or to look at drug combinations in vivo, to

 

 22   prioritize agents for Phase I trials, to make

 

 23   rational decisions within the pediatric consortia

 

 24   as to whether to continue to develop drugs or

 

 25   whether, at some point, we should drop those drugs

 

                                                               233

 

  1   in further development, preferably at the Phase I

 

  2   to Phase II transition to allow us to potentially

 

  3   focus a drug in treatment of certain tumors for the

 

  4   specific activity against certain models in the

 

  5   pediatric clinical screening program, and the

 

  6   potential to relate target inhibition to biological

 

  7   response which is going to become progressively

 

  8   more important as we deal with more agents that are

 

  9   inhibitors of specific signaling pathways.

 

 10             So, the data that suggests that some of

 

 11   these preclinical models may be useful is shown

 

 12   here.  This is from rhabdomyosarcoma models, and

 

 13   this is data that was gathered over about a 10-12

 

 14   year period in my own lab which identified

 

 15   vincristine, cytoxan, dactinomycin D and Adriamycin

 

 16   as having good activity against panels of

 

 17   rhabdomyosarcoma xenografts.  On the right column

 

 18   are sort of the response rates that have been

 

 19   gleaned from the literature that was available.

 

 20             On the other hand, it shows on the bottom

 

 21   that norfolan is a very active agent in the

 

 22   preclinical models and, indeed, is very active in

 

 23   rhabdomyosarcomas.  However, there is a cautionary

 

 24   note here.  Although we can identify drugs that are

 

 25   active in model systems and potentially active in

 

                                                               234

 

  1   the clinical setting, it doesn't necessarily mean

 

  2   that this is going to be a good drug.  The

 

  3   limitation of norfolan is that it causes cumulative

 

  4   toxicity to bone marrow and subsequently limits the

 

  5   ability to deliver standard therapy to those

 

  6   children.

 

  7             So, we have to look at these results as

 

  8   being promising in terms of being able to

 

  9   retrospectively identify drugs that we know are

 

 10   active in the clinical setting and to prospectively

 

 11   identify drugs that may have activity.  That

 

 12   doesn't necessarily mean to say that that drug is

 

 13   going to be potentially a very useful drug in the

 

 14   clinical setting.  So, there is a limitation to the

 

 15   models even though they are very promising.

 

 16   Ultimately, the value of the entity itself has to

 

 17   be determined in clinical trials.  We can merely

 

 18   point in that direction.

 

 19             On the other hand, you can take the same

 

 20   drugs and run those against colorectal

 

 21   adenocarcinoma xenografts, again, in

 

 22   immunodeficient mice and you see that the drugs

 

 23   that are very active against pediatric

 

 24   rhabdomyosarcoma essentially have no activity

 

 25   against the colon xenografts.  So, that gives you a

 

                                                               235

 

  1   little bit more confidence that it is not the fact

 

  2   that you have heterografted a tumor into a mouse

 

  3   that dictates its response.

 

  4             Coming to some more recent data, we have

 

  5   established a series of Wilms tumor xenografts, WT1

 

  6   through WT10, favorable histology Wilms tumors, and

 

  7   SKNEP is a cell line that was derived from a

 

  8   diffused xenoplastic Wilms tumor and the more

 

  9   pluses there are, the more sensitive the tumor is.

 

 10   So, anything that is greater than a 4-plus is an

 

 11   objective response in this model, so 50 percent

 

 12   regression in tumor size.

 

 13             So, you can see with vincristine, WT1

 

 14   through WT10, is 6-pluses which means that these

 

 15   tumors completely regress and do not regrow within

 

 16   a 12-week period of time.  Similarly, cytoxan has

 

 17   very good activity in most of the tumors.

 

 18   Prospectively, the model identifies the

 

 19   camphotecan, topotecan and irinotecan as being very

 

 20   active.  The important thing here is that topotecan

 

 21   and irinotecan are administered at doses that give

 

 22   relevant systemic exposures to humans.

 

 23             The relative exposure is perhaps the most

 

 24   important change in the way we are thinking about

 

 25   how to look at efficacy.  Efficacy in animal models

 

                                                               236

 

  1   is defined as the anti-tumor effect, let's say the

 

  2   ability of a drug to inhibit growth by 50 percent,

 

  3   divided by the dose that causes 10 percent

 

  4   lethality.  The problem is that the mouse is not a

 

  5   very good model for human toxicity.  The mouse may

 

  6   be either less tolerant to a drug, in which case

 

  7   you may under-predict the activity against a human

 

  8   tumor, or may be much more tolerant than a human,

 

  9   in which case the drug looks fantastic against the

 

 10   heterograft but ultimately fails in the clinic

 

 11   because you can't achieve systemic exposures in the

 

 12   patient that are consistent with the tumor

 

 13   regression in the mouse.

 

 14             The data shown here is the responses of

 

 15   different neuroblastoma xenografts to the drug

 

 16   topotecan against systemic exposure or area under

 

 17   the curve in nanograms per milliliter, showing that

 

 18   if we target, where the arrow is, 100 ng/ml we

 

 19   would expect to get in this case four out of the

 

 20   five tumor lines to give some response.  In fact,

 

 21   the total data set was the sixth line which is also

 

 22   completely resistant to topotecan.  So, we would

 

 23   predict if we targeted 100 ng/ml that we would have

 

 24   a response rate of four out of six or around 60

 

 25   percent.

 

                                                               237

 

  1             These sort of data are interesting but

 

  2   ultimately you have to prove or validate that this

 

  3   approach does have some merit.  That has been done

 

  4   in a clinical trial that was headed by Victor

 

  5   Santana, and the pharmacokinetics was done by

 

  6   Clinton Stuart, at St. Jude.  The idea here was to

 

  7   target the same systemic exposure that we set in

 

  8   the mouse, the 100 ng/ml of topotecan-lactone, and

 

  9   the design of the clinical trial is shown at the

 

 10   bottom.

 

 11             The drug is given for five days on two

 

 12   consecutive weeks, which is the schedule that is

 

 13   most effective in the xenograft models, and on day

 

 14   one there are pharmacokinetics taken and then the

 

 15   dose is adjusted to hit this target dose.  We are

 

 16   getting quite good at doing this.  In this

 

 17   particular trial there were 113 courses of drug

 

 18   administered and 92 percent were in the 100-plus or

 

 19   minus-20 ng/ml range.

 

 20             The results are shown here, where for 28

 

 21   evaluable patients we had approximately a 60

 

 22   percent response rate which is very close to that

 

 23   which would be predicted from a limited number of

 

 24   xenograft models, again suggesting that the idea of

 

 25   using pharmacokinetics as the metric against which

 

                                                               238

 

  1   anti-tumor activity is measured is perhaps more

 

  2   appropriate than using mouse toxicity per se.

 

  3             We looked at the retrospective analysis,

 

  4   again, about ten years work, and we see that for

 

  5   drugs that really didn't progress from Phase I any

 

  6   further, the area under the curve at the mouse

 

  7   maximum tolerated dose versus that in the human is

 

  8   somewhat higher in the mouse than the human.  So,

 

  9   in this example, the mouse is about 80 times more

 

 10   tolerant than are humans.  Yet, when one looks at

 

 11   the effective dose range, the effective dose range

 

 12   being reductions from the maximum tolerated dose in

 

 13   the mouse at which point you lose objective

 

 14   regressions in your tumor models, we see that the

 

 15   effective dose range for these drugs is relatively

 

 16   small, between two and three.

 

 17             So, where you have such discrepancy in the

 

 18   tolerance between the species and, yet, a very

 

 19   narrow window of true activity against the model

 

 20   systems which are human, one would anticipate these

 

 21   drugs would not necessarily achieve adequate

 

 22   concentrations to give tumor responses in a

 

 23   clinical situation.

 

 24             Drugs that work are shown here.  Norfolan,

 

 25   despite its limitations, is very active and, again,

 

                                                               239

 

  1   the pharmacokinetics in the mouse and the human in

 

  2   terms of tolerance are very similar.  There is a

 

  3   reasonable dose range of three to four before you

 

  4   lose activity.  Similarly, for topotecan the

 

  5   effective dose range spans the differential between

 

  6   mouse and human, as does irinotecan which is very

 

  7   well tolerated in terms of the active metabolite in

 

  8   the mouse and has an extremely wide therapeutic

 

  9   range within this model system.

 

 10             Looking at a more recent drug, irofulvin,

 

 11   NGAI114, again, anything that is more than a 4-plus

 

 12   is causing objective regressions.  We have looked

 

 13   at some 18 models.  It looks very active.  If you

 

 14   dose reduce you see that the MMT is somewhere

 

 15   between 4 and 7.  So, let's say you have 14 out of

 

 16   18 tumors, independent tumors show activity in

 

 17   terms of objective regressions.  As we reduce the

 

 18   dose further, it is 8 out of 18; reduce the dose

 

 19   further, it is 3 out of 15; and the lowest dose

 

 20   evaluated, only 1 out of 14 different tumors showed

 

 21   objective regressions.  These include tumors

 

 22   derived from brain tumors, neuroblastoma and

 

 23   rhabdomyosarcoma.

 

 24             The problem here is that even at this dose

 

 25   the systemic exposure to this drug in the mouse

 

                                                               240

 

  1   exceeds ten-fold that which can be achieved in

 

  2   human trials, again, suggesting that here is a drug

 

  3   that looks dramatically active in a model system

 

  4   but when you relate that activity to the ability to

 

  5   achieve systemic exposures of the drug in human it

 

  6   would suggest that this is a drug that would not be

 

  7   of high priority to undertake clinical trials, or

 

  8   at least progress from Phase I to Phase II clinical

 

  9   trials.

 

 10             Similarly, I think we can address issues

 

 11   of schedule-dependent anti-tumor activity.  This is

 

 12   old data with topotecan, but topotecan is given for

 

 13   5 days for every 21 days over 3 cycles, or given

 

 14   for 5 days times 2, so it is Monday through Friday;

 

 15   Monday through Friday at half the dose.  So the

 

 16   cumulative dose in both of these trials is exactly

 

 17   the same but the outcome in terms of tumor response

 

 18   is very different.

 

 19             I think this sort of data, where it is

 

 20   derived in a substantial number of tumor models to

 

 21   show that this is a consistent finding, may also be

 

 22   quite valuable in leading us in the design of

 

 23   clinical trials especially where, as was mentioned

 

 24   earlier today, one tends to get one shot at doing a

 

 25   large clinical trial and we might as well give it

 

                                                               241

 

  1   the best chance we can.

 

  2             The other aspect is the discrimination

 

  3   analogs of a particular class of chemical.  I think

 

  4   the models again can be quite useful if you apply

 

  5   this in the context of the achievable systemic

 

  6   exposures in humans versus the mouse.  This shows

 

  7   some data from an osteosarcoma xenograft which is

 

  8   particularly sensitive to carboplatin and

 

  9   cisplatinum but oxaliplatin, which is a drug that

 

 10   is of current interest in the pediatric oncology

 

 11   world, shows essentially no activity.  I think if

 

 12   one extends this data to, say, 6-10 osteosarcoma

 

 13   models and sees that, in fact, oxaliplatin has very

 

 14   little or no activity against these models, this

 

 15   can be factored into how we develop this drug in

 

 16   the clinical setting.

 

 17             These are classical cytotoxic drugs and,

 

 18   obviously, over the next few years there is going

 

 19   to be a progressive shift to drugs that we fondly

 

 20   call molecularly targeted drugs, even though

 

 21   perhaps they aren't quite as specific as we think

 

 22   they are.  But under those conditions we have to

 

 23   generate models that very accurately recapitulate

 

 24   the activity of, for example, signaling

 

 25   transduction pathways.  This raises the question of

 

                                                               242

 

  1   whether the conventional subcutaneous model is, in

 

  2   fact, going to be useful or whether we will have to

 

  3   go to models where the tumor is implanted into the

 

  4   more physiologically relevant sites, such as brain

 

  5   tumors into the brain, Wilms tumors into the

 

  6   kidney, etc.

 

  7             One way of addressing whether this is the

 

  8   case or not is through expression profiling and

 

  9   proteomics profiling, as alluded to by Paul

 

 10   Meltzer.  We have been looking at nearly

 

 11   established models and I will show you a couple of

 

 12   examples here where we have looked at Wilms tumors

 

 13   when we transplanted them into mice as xenografts

 

 14   and have done profiling from the primary tumor from

 

 15   which this xenograft was derived and the xenograft.

 

 16             So, what we are looking at here is the

 

 17   expression profiles for about 6,000 genes that are

 

 18   expressed at reasonable levels in the xenograft

 

 19   versus the primary tumor.  As you can see, there is

 

 20   a very high level of concordance, with about 20-30

 

 21   genes that are expressed greater than one standard

 

 22   deviation from the mean.  The data suggests very

 

 23   strongly that the expression profiles that are

 

 24   observed in the primary tumor are very largely

 

 25   recapitulated in the early xenograft studies.

 

                                                               243

 

  1             This means two things.  It gives us the

 

  2   first real metric to say this model is

 

  3   representative of the parental tumor because

 

  4   previously we have looked at histology and maybe

 

  5   measured a few antigens to see whether they are

 

  6   retained or not.  Now we can do this by profiling

 

  7   25,000 to 30,000 genes.

 

  8             Then having this data set, we can do two

 

  9   things.  One is, as these tumors are serially

 

 10   passaged in mice, from one mouse to another, we can

 

 11   ask a very pertinent question, at what point do

 

 12   these models start to deviate from the original

 

 13   tumor and, thus, may have much less relevance,

 

 14   particularly for screening or evaluating activity

 

 15   of molecularly targeted drugs.

 

 16             The second use is that if you have really

 

 17   consistent profiles like this, and these are

 

 18   maintained for multiple generations in the mouse,

 

 19   then we have the ability to look at the effects of

 

 20   drugs to perturb these profiles and start to get

 

 21   molecular signatures that may relate to biological

 

 22   outcome, that is, tumor response.

 

 23             One of the uses that we have made of this

 

 24   data is in collaboration with GlaxoSmithKline in

 

 25   cytokinetics, who had data, shown here, that the

 

                                                               244

 

  1   gene for the mitotic KSP was expressed at

 

  2   relatively high levels in tumors and particularly

 

  3   in Wilms tumor.  So, the arrow shows the levels of

 

  4   expression in normal kidney, which is extremely

 

  5   low, and also in clear cell carcinoma of the kidney

 

  6   and transitional cell carcinoma the expression of

 

  7   KSP is very low, but in Wilms tumor, which is

 

  8   circled here, it is extremely high.

 

  9             It allowed us to ask the question whether

 

 10   high levels of expression of KSP did, in fact, make

 

 11   this a drug target.  We have looked at one of the

 

 12   analogs of an anti-kinase inhibitor that is an

 

 13   analog of the compound that is currently in the

 

 14   clinic, and we have looked at this against a panel

 

 15   of Wilms tumors.  The bottom line is that this is a

 

 16   very active agent against favorable histology Wilms

 

 17   tumors that over-express KSP.  It is,

 

 18   unfortunately, not particularly useful against the

 

 19   diffuse anaplastic variety, here.  That is based

 

 20   upon a single xenograft and we are trying to

 

 21   establish further models and will see if that is,

 

 22   in fact, the case.

 

 23             In terms of the anti-tumor activity, if we

 

 24   can just focus here, this is tumor volume versus

 

 25   time after starting treatment.  Control is here. 

 

                                                               245

 

  1   This is the KSP inhibitor inducing complete

 

  2   regressions, with only 2 out of the 5 tumors

 

  3   regrowing during the 12-week period of observation.

 

  4             The limitation of this particular analog

 

  5   is that whilst it works very well at the highest

 

  6   dose, there is a very steep dose-response curve and

 

  7   there are much less active fractions of the MTD.

 

  8   So, again, this is going to be a drug where the

 

  9   relative pharmacokinetics between the mouse and the

 

 10   human are going to be really quite critical in

 

 11   determining whether this is very likely to have

 

 12   therapeutic benefit in these tumors.

 

 13             The final part of this is really this

 

 14   aspect of pharmacodynamics.  As all of us know, to

 

 15   look at target inhibition and, more specifically,

 

 16   target inhibition and target recovery in patients

 

 17   with solid tumors has been, and will remain to be,

 

 18   a very difficult proposition.  Multiple biopsies of

 

 19   tumor at various times before and after treatment

 

 20   is in most cases not really possible.

 

 21             I think the models can be quite useful in

 

 22   this respect and I will illustrate that in terms of

 

 23   the signaling pathway that we will be looking at in

 

 24   the context of a therapeutic trial of a rapamycin

 

 25   analog, CCI779.  This particular analog targets

 

                                                               246

 

  1   serene kinase, and it is very easy to monitor the

 

  2   effect of this drug by looking at downstream

 

  3   effectors and whether they are phosphorylated

 

  4   downstream.

 

  5             The problem is that target inhibition is

 

  6   only the first part of the question that you really

 

  7   want to ask.  That is, you are really asking at the

 

  8   drug doses that I am giving am I inhibiting the

 

  9   target?  That is the first part.  But what you

 

 10   really want to know is does the inhibition of

 

 11   target correlate with biologic readout.

 

 12             I think the model systems are going to be

 

 13   very useful to link the pharmacokinetics to target

 

 14   inhibition to biological readout in terms of

 

 15   anti-tumor activity, but even more so in terms of

 

 16   developing concepts of molecular signatures that

 

 17   may be much more important in predicting the

 

 18   outcome for treatment than merely looking at the

 

 19   target inhibition per se.

 

 20             Malcolm Smith will discuss this but the

 

 21   developing initiatives at the NCI include to

 

 22   systematically characterize tumors at the molecular

 

 23   level using both genomic and proteomic arrays.  The

 

 24   second is the Pediatric Preclinical Testing Program

 

 25   where we hope to establish models to identify new

 

                                                               247

 

  1   active drugs.

 

  2             I think in terms of using preclinical or

 

  3   nonclinical data we have to standardize our

 

  4   experimental procedures.  This is going to be

 

  5   difficult, but in the context of the proposed

 

  6   consortium that I have described it is difficult

 

  7   but it is a realistic goal, and I think once we

 

  8   have a group that is doing this on a large scale

 

  9   under consistent conditions, then I think others

 

 10   outside of that consortium who are doing similar

 

 11   work may adopt the same criteria for looking at

 

 12   tumor response and the design of experiments so

 

 13   that their data and our data can be compared and

 

 14   normalized.  I think we have to be careful that we

 

 15   use standardized criteria for assessing drug

 

 16   activity and, again, I think this is something that

 

 17   will come out of the consortium or the PPTP

 

 18   initiative, whoever carries that out.

 

 19             One of the other questions that was being

 

 20   raised is should we be using animal data that is

 

 21   derived under Good Laboratory Practice compliance.

 

 22   The problem here is that if we do this for the

 

 23   cancer screening program, then my understanding is

 

 24   that the entire vivarium within an institute or

 

 25   university also has to function under GLP

 

                                                               248

 

  1   conditions and this aspect of the work may be a

 

  2   very small percentage of the total work that is

 

  3   being done in a vivarium per se.  It would

 

  4   certainly increase the costs quite dramatically so

 

  5   I think we have to think about the prospect of GLP

 

  6   in the context of who is going to be doing this

 

  7   work and whether this would increase the cost of

 

  8   animal experimentation not only for the work that

 

  9   is being focused on cancer, but also for non-cancer

 

 10   related work that is ongoing in the same

 

 11   institution.  Thank you.

 

 12             DR. SANTANA:  Thank you, Peter for a very

 

 13   thorough overview of this issue.  I am going to ask

 

 14   Chand Khanna to go ahead and do his presentation.

 

 15   After that we will take a break and then we will

 

 16   come back and reconvene and finish the last two

 

 17   presentations and have our discussion and

 

 18   questions.

 

 19            An Integrated and Comparative Approach to

 

 20              Preclinical/Clinical Drug Development

 

 21             DR. KHANNA:  I want to thank everyone for

 

 22   the opportunity, specifically Steven, to come and

 

 23   speak to you today.

 

 24             As Peter suggested, the convention to drug

 

 25   development, as you all know, is to include

 

                                                               249

 

  1   preclinical models to evaluate promising agents and

 

  2   then move those promising agents through clinical

 

  3   development.  To continue Peter's theme, what I

 

  4   would like to present is a vision towards an

 

  5   integrated approach wherein preclinical models can

 

  6   be helpful and informative, both at the preclinical

 

  7   level and during various phases of clinical

 

  8   development, and the spin that I would like to

 

  9   provide is one that includes a number of novel

 

 10   models, models that have not been used very much in

 

 11   drug development, and those include naturally

 

 12   occurring cancers that are seen in both genetically

 

 13   engineered mice and, more specifically, in pet

 

 14   animals in our communities that can, again, be

 

 15   included in translational and biological cancer

 

 16   research.

 

 17             What I am going to do is to bring this to

 

 18   you from my efforts within the Comparative Oncology

 

 19   Program of the CNI, which is a new initiative

 

 20   within the Center for Cancer Research, and my work

 

 21   with the Pediatric Oncology Branch where my focus

 

 22   is on sarcoma biology and metastasis.

 

 23             As Peter has alluded to, there are a

 

 24   number of modeling options, and the ones he has

 

 25   focused on and shown us really are how we can best

 

                                                               250

 

  1   use the xenograft models, but there are also

 

  2   opportunities for us to include syngeneic or mouse

 

  3   cancers that are transplantable into mice, and

 

  4   genetically engineered mice that can be used for a

 

  5   number of important steps in the translational

 

  6   process.  Lastly, what I want to focus on is the

 

  7   use of pet animals in the drug development process.

 

  8             This is a schema that you are familiar

 

  9   with, wherein small animals are used early in the

 

 10   development.  Primarily for toxicology we use large

 

 11   animals, whether they be non-human primates or

 

 12   dogs, and then we move into clinical development.

 

 13   The question is how can we use first a small set of

 

 14   examples genetically engineered mice to inform this

 

 15   process?  Largely, I think, because they are more

 

 16   complicated and challenging, we can use them in the

 

 17   evaluation of interesting findings from traditional

 

 18   transplantation models.

 

 19             So, if we look at the historical

 

 20   perspective, genetically engineered mice have been

 

 21   problematic for basically three primary reasons:

 

 22   One is that they are conventionally associated with

 

 23   very rapid tumor progression.  They are

 

 24   historically associated with hemologic malignancies

 

 25   and the cancers that emerge usually emerge in a

 

                                                               251

 

  1   number of sites synchronously.

 

  2             Recently there have been novel modeling

 

  3   approaches which have provided us an opportunity to

 

  4   study genetically engineered mice across a range of

 

  5   cancer histologies, almost all histologies.

 

  6   Through efforts including conditional expression of

 

  7   genes, somatic expression of genes within a

 

  8   selected pool of target cells, there are now very

 

  9   good mouse models for most human cancers.  The

 

 10   advantages that these genetically engineered mice

 

 11   provide through the translational process are that

 

 12   after you induce the genetic change in the mouse

 

 13   the cancers that emerge, emerge spontaneously.

 

 14   That is one.

 

 15             The second is that the tumor that emerges

 

 16   is syngeneic from the tumor to the tumor

 

 17   micro-environment to the host, and that is

 

 18   something that I think provides opportunities

 

 19   specifically for targeted biology-based therapies.

 

 20   The genetics of the cancer are modifiable and are

 

 21   relevant and, although it is more easily said than

 

 22   done, the biology of these cancers can be

 

 23   controlled now so we can have opportunities for

 

 24   therapeutic evaluation during the course of

 

 25   progression that these genetically engineered mice

 

                                                               252

 

  1   have.

 

  2             There are limitations, and the limitations

 

  3   that we see with traditional animals still exist

 

  4   with these genetically engineered mice.  There is

 

  5   heterogeneity within a specific population of mice.

 

  6   There is heterogeneity in the genetics of the

 

  7   cancer and I think that is a value.  It adds to

 

  8   what we get out of more or less homogeneous

 

  9   populations seen in the transplantation settings.

 

 10   Experimentally, these are very difficult and

 

 11   complicated designs to pursue from the standpoint

 

 12   of translation but they can be done.  They are

 

 13   expensive, time consuming, and we don't really know

 

 14   yet about their predictivity.

 

 15             The most important issue about their use

 

 16   is a series of patents that have been provided to

 

 17   Dupont exclusively that really extend to all

 

 18   genetically engineered mice.  Any activated

 

 19   oncogene in a mouse is covered by the OncoMouse

 

 20   patents.  The result of these patents is really the

 

 21   limitation of their use in the pharmaceutical

 

 22   industry.  So, unless this issue can be dealt with,

 

 23   I think the use of these genetically engineered

 

 24   mice in the pharmaceutical industry will be

 

 25   limited.

 

                                                               253

 

  1             What I want to move on to is ways for us

 

  2   to include naturally occurring cancers in the

 

  3   translational process in the drug development

 

  4   process.  Again, within the Comparative Oncology

 

  5   Program what we plan to provide are opportunities

 

  6   to include these models in drug development.  So,

 

  7   pet animals have a number of interesting cancers

 

  8   that are relevant from the standpoint of pediatric

 

  9   cancers, including lymphoma and then dogs with

 

 10   osteogenic sarcoma.

 

 11             Dogs in the community are developing these

 

 12   cancers.  There are 65 million pet dogs in the

 

 13   United States, 6 million will develop cancer in a

 

 14   year and the pet owners of these dogs are seeking

 

 15   out advanced care and, in many cases, are very

 

 16   interested in including their dogs in trials that

 

 17   evaluate new therapies.  So, what this provides is

 

 18   an opportunity to include these large animal models

 

 19   in drug development and this has been done largely

 

 20   within the pharmaceutical industry.

 

 21             The advantage that these large animals

 

 22   provide is, in fact, that they are large outbred

 

 23   animals, unlike the small animals that we

 

 24   traditionally use at the preclinical level.  The

 

 25   genetics of the host, the dogs, have been shown by

 

                                                               254

 

  1   the recent completion of the canine genome to be

 

  2   quite similar, very similar in fact, to humans.

 

  3   They are naturally occurring cancers.  Then, within

 

  4   given histologies the genetics of the cancers are

 

  5   very similar to the genetics of the same human

 

  6   cancers.  Very importantly, one thing that these

 

  7   models provide is that within a histology there is

 

  8   considerable genetic and individual variability

 

  9   that is, in fact, captured within populations of

 

 10   humans and often is the problem as we move through

 

 11   clinical development.  This heterogeneity is not

 

 12   captured in other models.

 

 13             If you look at histology responses, for

 

 14   example  lymphoma, the drugs that are effective in

 

 15   dogs with lymphoma are effective in people with

 

 16   lymphoma.  The drugs that are not effective in dogs

 

 17   with lymphoma are not effective in people with

 

 18   lymphoma.  To a large extent, that parallel is true

 

 19   for a number of histologies with classical,

 

 20   conventional cytotoxic drugs.  The biology of

 

 21   metastases within these models is faithfully

 

 22   reproduced for specific histologies.  Lastly, I

 

 23   think an important point is that these cancers are

 

 24   characterized by resistance or recurrence and this

 

 25   is really the problem that we face with pediatric

 

                                                               255

 

  1   patients and adult patients.  The biology of

 

  2   recurrence or resistance is difficult to model in

 

  3   most small animal settings.

 

  4             So, if we look at this table that I have

 

  5   taken from Shadner's recent review in JCO, he has

 

  6   listed out preclinical through clinical development

 

  7   of the number of agents at various phases at one

 

  8   point in time.  What I have done in red is just put

 

  9   the number of agents that are active per year.  By

 

 10   looking at this, you can see where opportunities

 

 11   exist to improve the process of drug development.

 

 12   Certainly as Peter suggested, there is room for us

 

 13   to improve this initial step but as we move along,

 

 14   I think there are great opportunities for us to

 

 15   take Phase I agents that are not burdened by the

 

 16   hurdle of maximally tolerated dose and inform

 

 17   decisions towards Phase II.  I think there is an

 

 18   opportunity for these large animal models, for

 

 19   genetically engineered mice to take that role of

 

 20   informing towards Phase II and potentially

 

 21   informing towards Phase III.

 

 22             So, this is the integrated approach that I

 

 23   would like to suggest wherein pet dogs--we have

 

 24   largely done this work within the pharmaceutical

 

 25   industry to assess activity, toxicity,

 

                                                               256

 

  1   pharmacokinetics and pharmacodynamics and used that

 

  2   information to lead towards Phase I.  Well, perhaps

 

  3   as important, use these tumor-bearing dog studies

 

  4   to define dose regimen schedules towards Phase II

 

  5   to validate, potentially to identify but really

 

  6   more appropriately validate biomarkers, define

 

  7   responding histologies, and then provide a rational

 

  8   system in which we can demonstrate that

 

  9   combinations should be considered towards Phase II

 

 10   and potentially Phase III.

 

 11             So, I would like to give you a couple of

 

 12   short examples.  Thrombospondin-1 is a very large

 

 13   protein with a number of receptors and a number of

 

 14   effector domains.  The second type-1 repeat has

 

 15   been associated with significant antiangiogenic

 

 16   activity.  From the second type-1 repeat a series

 

 17   of small peptides, non-amino acid peptides, are

 

 18   being pursued as anti-cancer drugs, antiangiogenic

 

 19   drugs.  The problem with the development of this

 

 20   class of drugs and specifically thrombospondin-1 is

 

 21   that although we can show within mice that these

 

 22   agents are antiangiogenic and although we can show

 

 23   that they do have anti-cancer activity, the leap

 

 24   towards the clinic has been difficult.

 

 25             So, the question was whether or not we

 

                                                               257

 

  1   could use dogs with naturally occurring cancers to

 

  2   help us make that step.  What I would like to show

 

  3   you is a simple example of how we have done that.

 

  4   The experimental clinical trial for pet dogs

 

  5   included dogs with any measurable malignant cancer,

 

  6   no concurrent therapy, and the endpoints really

 

  7   were to assess toxicity, a limited attempt to

 

  8   evaluate PK, and then to look at response, keeping

 

  9   in mind that response was going to be assessed

 

 10   against bulky disease using a single-agent

 

 11   antiangiogenic drug.

 

 12             The first point that I want to bring up is

 

 13   that accrual is achievable.  In a short period of

 

 14   time we can enter large numbers of dogs in these

 

 15   clinical trials with the support and interest of

 

 16   their pet owners.  Toxicity has always been

 

 17   evaluated, in fact, in dogs.  An interesting and

 

 18   important point is that pet dogs that bear cancer

 

 19   have different toxicity profiles than beagle dogs

 

 20   that are evaluated in the research setting.  In

 

 21   fact, in many situations the toxicities that are

 

 22   seen in pet dogs are much more similar to those

 

 23   toxicities seen in patient populations.

 

 24             I will show you some of the responses.

 

 25   This is a dog with a maxillary squamous cell

 

                                                               258

 

  1   carcinoma.  This is the lesion after 30 days on

 

  2   therapy.  It is perhaps a little clearer here.

 

  3   After 60 days the lesion is much more active.  It

 

  4   is hemorrhagic.  Through a 60-day period of time in

 

  5   a human clinical trial, Phase I trial, it is

 

  6   unlikely that you would continue this patient on

 

  7   therapy with progression.  But we did continue this

 

  8   dog and after 90 days, the lesion is now no longer

 

  9   present.  We can biopsy this site and there is

 

 10   squamous cell carcinoma that is persistent there

 

 11   but the lesion is not actually assessable there.

 

 12   So, this dog continues to do well, free of disease

 

 13   that is measurable within the mouth, but not a

 

 14   histological regression.

 

 15             I have several other images that I could

 

 16   show you to suggest, in fact, that the agents are

 

 17   active and they can result in regressions.  The

 

 18   responses include stabilization which we feel are

 

 19   real but, in fact, objective regressions of lesions

 

 20   that cross a number of histologies.

 

 21             The other thing that this points to is, in

 

 22   fact, histologies that we wouldn't have predicted

 

 23   activity in. So, lymphoma was found to be quite an

 

 24   active site and now, in Phase II, these drugs are

 

 25   moving ahead.  Of interest to the group, sarcomas

 

                                                               259

 

  1   were particularly responsive histology.

 

  2             So, what did we learn from these dog

 

  3   studies?  Antiangiogenic peptides can be active

 

  4   against bulky disease.  They need time.  Because of

 

  5   the results that we were able to generate in dogs,

 

  6   the Phase I trials in Europe extended their

 

  7   observation times and they did see objective

 

  8   responses in patients treated for 60 days.

 

  9             Agents are active against histologies we

 

 10   wouldn't have predicted, like non-Hodgkin's

 

 11   lymphoma.  A very important point is that all dogs

 

 12   that continue through therapy develop resistance on

 

 13   therapy so combinations are going to be necessary

 

 14   and, as we look towards the use of these agents, we

 

 15   are going to have to keep in mind that resistance

 

 16   will be an obvious problem.  Most dogs don't

 

 17   respond to therapy and, therefore, there is an

 

 18   opportunity for us to define markers that predict

 

 19   responsiveness within a heterogeneous population of

 

 20   dogs and, in fact, predict when responses will be

 

 21   seen.  That work is being done and thus far

 

 22   circulating endothelial cells seem to an interest

 

 23   and will move on into the clinical setting as well.

 

 24             This is, again, the perspective that we

 

 25   have and I think there are some examples from the

 

                                                               260

 

  1   thrombospondin-1 studies that show how we can

 

  2   inform towards Phase II.  I want to end with

 

  3   another brief example and it speaks to this

 

  4   pharmacokinetic/pharmacodynamic response question

 

  5   that Peter brought up.

 

  6             So, Cheryl London, who is at UC Davis, is

 

  7   evaluating small molecule inhibitors of the split

 

  8   tyrosine kinase receptor family.  What she was able

 

  9   to do in a very similar trial design, treating dogs

 

 10   with bulky disease, is actually do tumor

 

 11   pharmacodynamics using phospyl KiT as the target;

 

 12   do serial biopsies in dogs evaluating the diversity

 

 13   of KiT mutations in dogs with nasal tumors and

 

 14   define the dose that is required to modulate the

 

 15   target in vivo to validate surrogates that could be

 

 16   more evaluated in human clinical populations

 

 17   against this tumor target, and then move those

 

 18   things into the clinic.

 

 19             She was able to show that the dosing

 

 20   schedule, an every other day dosing schedule, was

 

 21   valuable and able to achieve threshold receptor

 

 22   inhibition of KiT.  This information was translated

 

 23   directly into the development of products in

 

 24   clinical trials.  The every other day dose was

 

 25   suggested for human development but the human

 

                                                               261

 

  1   development required input from marketing and

 

  2   marketing didn't want to pursue every other day

 

  3   dosing.  The drug trials predicted daily dosing

 

  4   would be toxic and, in fact, was toxic in people.

 

  5             I am just going to jump ahead.  So, what

 

  6   we are interested in being able to do within the

 

  7   Comparative Oncology Program is provide a reagent

 

  8   kit that can allow biology-based questions to be

 

  9   answered in these trials.  This has been a

 

 10   difficulty for dog trials thus far in that we just

 

 11   don't have reagents to study dogs in a rigorous

 

 12   way.  We now have a validated canine oligoarray, a

 

 13   17K element array.  We are validating proteomics

 

 14   approaches in dogs with cell signaling.  We have

 

 15   screened specific antibodies for cross-reactivity

 

 16   to dogs and we have made good progress there.

 

 17             Multicenter collaborations are going to be

 

 18   required for us to be able to do trials in a short

 

 19   period of time, and allow that short period of time

 

 20   to inform towards clinical development of the same

 

 21   drugs, and to be able to help with decisions of

 

 22   when these models can be used and when they should

 

 23   not be used in development.  There are times where

 

 24   really the questions are not appropriate to ask

 

 25   within these dog studies.

 

                                                               262

 

  1             I will just end with a list of histologies

 

  2   that I think are relevant.  Osteosarcoma is

 

  3   obviously an area of personal interest and we have

 

  4   actually published randomized, prospective,

 

  5   placebo-blinded trials in dogs with osteosarcoma

 

  6   looking for opportunities in the clinic.

 

  7             We are interested in lymphoma.  There are

 

  8   other histologies.  But important to note is that

 

  9   within each of these cancer histologies are genetic

 

 10   changes that can be modeled and can be targeted.

 

 11   So, it doesn't have to be histology based.

 

 12             The weaknesses of these models are the

 

 13   cost.  Drug costs are a primary concern; the cost

 

 14   of managing the trials and time.  They are longer

 

 15   models than what we would see with typical small

 

 16   animal studies although the time is much shorter

 

 17   than what you would have in the same clinical study

 

 18   in a human population.

 

 19             With that, I will conclude.  I will

 

 20   acknowledge our initial group in Comparative

 

 21   Oncology, and the slide also includes Lee Helman

 

 22   and the people in the Pediatric Oncology Branch.

 

 23   UC Davis and Cheryl London has been doing a lot of

 

 24   these translational studies.  Now, with the

 

 25   interest of CTEP and the CCR, we are pursuing some

 

                                                               263

 

  1   trials with 17DMAG to answer some of these

 

  2   questions that will inform towards Phase II.

 

  3             DR. SANTANA:  Thank you, Chand.  I will

 

  4   seek the advice of the FDA.  Should we take a

 

  5   ten-minute break and try to get back on schedule

 

  6   because I know we are going to have people dropping

 

  7   off as the day progresses.  So, why don't we just

 

  8   take a ten-minute break and reconvene at 3:00,

 

  9   finish with the two presentations and then take

 

 10   questions and discussion and try to get out of here

 

 11   on time?

 

 12             [Brief recess]

 

 13             DR. SANTANA:  I will invite our next

 

 14   speaker to come to the podium.  Dr. Kenneth

 

 15   Hastings will address the issues of what can be

 

 16   learned about safety using different models.

 

 17                  What can Learned About Safety?

 

 18             DR. HASTINGS:  Well, my task, after these

 

 19   really nice scientific presentations, is to give

 

 20   you the regulatory spin on things so your task is

 

 21   to stay awake.

 

 22             What I want to talk about today is the use

 

 23   of neonatal and juvenile animal studies for

 

 24   determining the safety of drugs for use in

 

 25   pediatric patients and, obviously, this is going to

 

                                                               264

 

  1   apply to pediatric oncology.

 

  2             The specific guidance that really led to

 

  3   the development of guidance on juvenile animal

 

  4   studies was the Pediatric Exclusivity Act under

 

  5   Section 505A of the FDC Act.  The specific language

 

  6   that is included that refers to nonclinical studies

 

  7   is that the FDA may request nonclinical trials

 

  8   before completing pediatric studies in humans.

 

  9   Certain toxicology studies in immature animals may

 

 10   be necessary to evaluate the safety of use in

 

 11   pediatric conditions.

 

 12             Also another regulatory background

 

 13   document has been referred to previously, and that

 

 14   is ICH E11, clinical investigation of medicinal

 

 15   products in the pediatric population, and once

 

 16   again the decision to proceed with a pediatric

 

 17   development program involves consideration of many

 

 18   factors, including any nonclinical safety issues.

 

 19   The need for juvenile animal studies should be

 

 20   considered on a case-by-case basis.  Then it refers

 

 21   to ICH M3, which is the document that outlines the

 

 22   timing of nonclinical studies vis-a-vis clinical

 

 23   studies.

 

 24             Finally, there is a draft document that

 

 25   was published in February, 2003, nonclinical safety

 

                                                               265

 

  1   evaluation of pediatric drug products.  We now have

 

  2   the final version, after comments were made to the

 

  3   docket, and we hope to publish it sometime this

 

  4   spring or summer, and we took into consideration

 

  5   the comments that were made.  This document

 

  6   provides guidance on the role and timing of animal

 

  7   studies in the safety evaluation of therapeutics

 

  8   intended for the treatment of pediatric patients,

 

  9   and it also provides specific recommendations based

 

 10   on the available science and pragmatic

 

 11   considerations.

 

 12             Why did we get into the issue of juvenile

 

 13   animal studies?  Well, in assessing the use of

 

 14   drugs for pediatric use the basic assumption that

 

 15   we have proceeded with over the years has been that

 

 16   under most circumstances the safety and efficacy of

 

 17   drugs approved for use in adults predicts pediatric

 

 18   use if you make the appropriate dose adjustment.

 

 19             Now, in the past we have used things like

 

 20   relative body surface area.  We consider that to be

 

 21   a good default measure for dose adjustment.  But

 

 22   generally this is less informative than data you

 

 23   would get from a clinical pharmacology study.  That

 

 24   is really what we are after, being able to make

 

 25   dose recommendations based on actual ADME

 

                                                               266

 

  1   pharmacokinetic studies.

 

  2             Neonatal and juvenile animal studies to

 

  3   enable clinical studies are needed basically to

 

  4   support the safety of studies in pediatric

 

  5   patients.  The origin of the guidance really was to

 

  6   provide information and what we call triggers on

 

  7   the need for nonclinical studies.  Basically, what

 

  8   we are saying here is that you don't need to do a

 

  9   juvenile animal study every time you want to do a

 

 10   clinical trial in a pediatric patient population.

 

 11   What we were trying to do is to find out what are

 

 12   the sorts of things that we could observe or

 

 13   already know about the toxicology or the safety of

 

 14   a drug that would tell us that maybe you need to do

 

 15   a pediatric juvenile animal study to support the

 

 16   safety of a pediatric study.

 

 17             Also, this guidance contains advice on the

 

 18   conduct of the studies and provides information on

 

 19   how the results of these studies would be used in

 

 20   designing pediatric drug trials and, in fact, in

 

 21   deciding whether or not they would be safe.

 

 22             Now, we recognize that there are

 

 23   differences in the drug safety profiles between

 

 24   mature and immature systems, and these include

 

 25   differences in susceptibility to insult and

 

                                                               267

 

  1   differences in toxicity-related ADME parameters.

 

  2   We recognize that some physiological systems are

 

  3   more vulnerable than others, especially those that

 

  4   undergo extensive postnatal development.

 

  5             When you think about it, you know, that

 

  6   doesn't exclude much.  There are a lot of things

 

  7   that undergo significant postnatal development.

 

  8   So, really more than anything else what we would

 

  9   think about are those that might be particularly

 

 10   susceptible to insult, such as the developing

 

 11   nervous system, maybe the developing immune system,

 

 12   the kidneys, perhaps even the gut.  So, those would

 

 13   be potential triggers for asking for a juvenile

 

 14   animal study if we knew from adults, from clinical

 

 15   practice or from mature animal studies, that these

 

 16   are target organs of toxicity.

 

 17             Now, I want you to keep in mind two basic

 

 18   concepts that toxicologists use all the time.  They

 

 19   have to do with how you look at the usefulness of

 

 20   studies, what it is that you intend to get out of

 

 21   the study.  Actually, I have them in reverse order.

 

 22   The first are studies that are designed for hazard

 

 23   identification.  Basically, the idea behind hazard

 

 24   identification is that you demonstrate that a drug

 

 25   or a candidate drug has the potential to cause an

 

                                                               268

 

  1   adverse effect.  An example of hazard

 

  2   identification would be something like an Ames

 

  3   assay or a discovery toxicology study where you

 

  4   administer a drug by intraperitoneal injection.

 

  5   You are just trying to find out if a drug can cause

 

  6   a toxicity.

 

  7             Pertinent to our discussion today, under

 

  8   certain circumstances adverse effects in mature

 

  9   animals might not be predictive of adverse effects

 

 10   in developing systems.  So, some studies that you

 

 11   might conduct, some juvenile animal studies you

 

 12   might conduct actually might be for the purposes of

 

 13   hazard identification, and I am going to talk about

 

 14   how that plays into the design of studies a little

 

 15   bit later.

 

 16             Risk assessment, of course, is that you

 

 17   are trying to look at all of the parameters of

 

 18   toxicity--systemic exposure, route of

 

 19   administration, length of exposure, all of the

 

 20   parameters that determine whether or not what is a

 

 21   potential toxicity is actually going to be manifest

 

 22   as a toxicity in the use of the drug.  Basically,

 

 23   this is one of the assumptions that we make when we

 

 24   say that for studies conducted in mature animals

 

 25   the effects will predict what happens in neonates. 

 

                                                               269

 

  1   What you need to do is determine what parameters,

 

  2   particularly ADME parameters might alter that risk.

 

  3             I want to just mention very briefly the

 

  4   differences in pediatric versus adult patients or

 

  5   subjects with respect to ADME because that really

 

  6   was the driving factor in looking at juvenile

 

  7   animal studies to start out with.  In humans, if

 

  8   you look at ADME, there are differences with age as

 

  9   far as distribution of drug dose.  The receptors

 

 10   come and go; they develop and certain

 

 11   age-restricted ranges and, therefore, what you

 

 12   observe in younger systems may not be applicable to

 

 13   older animals and, obviously, extrapolating this

 

 14   clinically.

 

 15             As far as absorption of an orally

 

 16   administered drug, you have to consider that in

 

 17   infants they have a larger volume of distribution,

 

 18   larger surface area to body weight ratio, and the

 

 19   body composition is different.  Infants and

 

 20   children have higher gastric pH which will affect

 

 21   the absorption of basic and acidic drugs, larger

 

 22   absorption of the basic drugs; less absorption of

 

 23   acidic drugs.  GI motility is different.  In

 

 24   infants and neonates GI motility tends to be fairly

 

 25   low compared to adults.  In children the motility

 

                                                               270

 

  1   tends to be high compared to adults.  So, the

 

  2   actual achievable AUC for a particular orally

 

  3   administered drug may be different if you just do

 

  4   your extrapolation based on body surface area.

 

  5   And, there are certain other things to consider,

 

  6   such as unique routes of exposure such as through

 

  7   mother's milk.

 

  8             A very difficult issue is metabolism.  We

 

  9   know that as a general rule there are certain

 

 10   metabolic systems that appear to be more functional

 

 11   in pediatric patients versus adults.  I am not

 

 12   going to get into a long discussion about

 

 13   differences in metabolism except to say this, with

 

 14   respect to P450 enzymes, if you look at juvenile

 

 15   animals and if you look particularly at rats which

 

 16   is a model that we use quite often, we actually

 

 17   don't know a lot about the relative development of

 

 18   the P450 enzymes.  There is probably one exception

 

 19   to that.  We thought that there would probably be a

 

 20   lot of information on this.  It turns out that

 

 21   actually there is not in the published literature.

 

 22             Finally, another thing to consider is

 

 23   excretion in juvenile animals--actually, I am

 

 24   talking clinically but in children you have lower

 

 25   glomerular filtration rate, lower tubular

 

                                                               271

 

  1   secretion, resulting in slower clearance and longer

 

  2   half-life.  Once you get up into the child range

 

  3   you have rapid clearance and shorter half-lives.

 

  4   So, once again, pharmacokinetics may not be

 

  5   predictable based on body surface area.

 

  6             One thing to consider is how valuable are

 

  7   animal models for ADME comparisons.  Well, an

 

  8   obvious advantage is that in animals you can do

 

  9   experimental manipulations that might help you

 

 10   define ADME parameters.  But a not so obvious

 

 11   advantage, as I have mentioned, is the lack of

 

 12   comparative information in animals, particularly

 

 13   with respect to metabolizing enzymes.  One thing to

 

 14   consider though is that if you can associate PK

 

 15   parameters with adverse effects in animals, this

 

 16   might be useful in clinical trials.  So, that is

 

 17   one real advantage to a juvenile animal model.

 

 18             Really ADME was what originally drove the

 

 19   consideration of doing juvenile animal studies.

 

 20   Obviously, the other thing that we are interested

 

 21   in is toxicity.  Are these studies going to be

 

 22   safe?  The things that we need to consider are the

 

 23   relative maturations of physiologic systems.  These

 

 24   are probably better understood in animals but we

 

 25   could have a debate about that.  If adverse effects

 

                                                               272

 

  1   are observed in mature animals, then the juvenile

 

  2   animals could be used to demonstrate increased or

 

  3   decreased susceptibility, and you may be able to

 

  4   understand how ADME might affect that.  Once again,

 

  5   however, extrapolation to clinical trials may be

 

  6   less certain because of the variations in, for

 

  7   instance, metabolism that we don't really

 

  8   understand as well as we should in animals.

 

  9             Let me lay out a couple of scenarios where

 

 10   juvenile animal studies might be useful for the

 

 11   purposes of toxicology studies.  One thing, you may

 

 12   need a juvenile animal study if you already have a

 

 13   pretty good handle on the adverse effects and you

 

 14   have a pretty good idea about the ratio of toxic

 

 15   dose to efficacious dose, and particularly this may

 

 16   be true for short-term use drugs like antibiotics.

 

 17             However, and this was mentioned earlier--I

 

 18   believe Dr. Santana mentioned this, sometimes even

 

 19   with acute exposure you might need long-term

 

 20   follow-up studies.  The classic example for this is

 

 21   the fluoroquinolones.  What happened here, as you

 

 22   probably are aware, fluoroquinolones are associated

 

 23   with a very troubling effect called crippling

 

 24   arthropathy.  It was originally discovered or

 

 25   described in puppies, in young dogs.  The question

 

                                                               273

 

  1   was the clinical relevance of these studies.  There

 

  2   is a lot of talk about this and I don't want to get

 

  3   into that debate but I think that most people

 

  4   nowadays consider that fluoroquinolone use in

 

  5   children is something you approach very carefully

 

  6   because this may very well be a serious adverse

 

  7   effect that would persist into adulthood.

 

  8             One of the ways that we have looked at

 

  9   answering this question was simply to do this, to

 

 10   dose juvenile dogs, beagles, with fluoroquinolones

 

 11   over a course of, like, two weeks at, say, doses

 

 12   equivalent or maybe higher than what you would use

 

 13   clinically, producing AUCs equivalent to higher

 

 14   than clinical doses, and then just let the dogs go,

 

 15   let them mature and then, at about six months of

 

 16   age, you would look at the dogs again and do

 

 17   clinical evaluations, to histopath on the affected

 

 18   bones and see if there are any changes in those

 

 19   animals; see if the effect gets worse; see if it

 

 20   improves; see if there are any associated lesions

 

 21   that appear to be caused by this juvenile exposure.

 

 22             In fact, what we now know about

 

 23   fluoroquinolones--to cut to the chase--is that

 

 24   actually these effects tend to persist.  They

 

 25   probably don't get worse but they do persist.  That

 

                                                               274

 

  1   is an important thing to learn in deciding whether

 

  2   or not to conduct a clinical trial, let's say, for

 

  3   something like otitis media, and also to look at

 

  4   the follow-up.  In fact, that was used as an

 

  5   argument for the long-term follow-up of children in

 

  6   clinical trials with fluoroquinolones, and I think

 

  7   this is something you should take into

 

  8   consideration when you think about oncolytics used

 

  9   in pediatric patients.  I think it is a pretty good

 

 10   comparison that you might want to think about.

 

 11             When you talk about long-term use,

 

 12   particularly, let's say, a drug that has never been

 

 13   developed for use in adults, then you might think

 

 14   about what we would call a shift to a hazard

 

 15   identification type of study.  What you would do

 

 16   here is you would start with juvenile animals.  You

 

 17   would dose them all the way through adulthood, look

 

 18   for adverse effects and then, if you do see adverse

 

 19   effects, you can go back and do window of

 

 20   vulnerability studies where you try to find out

 

 21   where, in the development of that animal, this

 

 22   occurred and this could help you in understanding

 

 23   where the vulnerable windows would be in a clinical

 

 24   trial.  In other words, you can build risk

 

 25   assessment into what is in fact, when you think

 

                                                               275

 

  1   about it conceptually, a hazard identification kind

 

  2   of study.  You can also build in pharmacokinetics,

 

  3   obviously, and safety pharmacology studies such as

 

  4   effects on blood pressure, cardiac function, renal

 

  5   function and things like that.

 

  6             I just want to make one mention about

 

  7   efficacy models.  We have had a lot of talk about

 

  8   efficacy models; very good talks.  I just want to

 

  9   say that you can build safety determinations into

 

 10   efficacy models, particularly large animal models

 

 11   where you can do serial blood levels of biomarkers

 

 12   or AUC for the drug, things like that.  So,

 

 13   although we haven't in the past typically looked at

 

 14   efficacy models for safety information--we do our

 

 15   toxicology studies in otherwise health animals,

 

 16   efficacy models probably can be used for this, and

 

 17   I think there is at least some experience with that

 

 18   in looking at biologics.

 

 19             Now I am going to mention the animal rule.

 

 20   The animal rule was passed, I believe, in 2001.  I

 

 21   think that is when it was finally codified.  This

 

 22   allows for use of animal studies to demonstrate

 

 23   efficacy for where clinical trials would be

 

 24   unethical and/or not feasible.  It applies to new

 

 25   drug and biologic products.  It is used to reduce

 

                                                               276

 

  1   or prevent toxicity of chemical, biological,

 

  2   radiological or nuclear substances.  Obviously, I

 

  3   think we can sort of see what the animal rule is

 

  4   really designed for, and that was for development

 

  5   of drugs to treat things like anthrax.  Basically,

 

  6   we are talking about counter-terrorism measures.

 

  7   You know, antidotes for nerve toxins and things

 

  8   like that.  That is what it is really designed for.

 

  9             Drugs considered should have demonstrated

 

 10   safety in humans.  That is one thing that is built

 

 11   into the animal rule.  Now, whether or not that

 

 12   would apply to oncolytics, that is a different

 

 13   question and I think that is something for the

 

 14   panel to discuss.  If possible, clinical activity

 

 15   in a relevant disease, although lack of clinical

 

 16   efficacy data shouldn't prejudice against

 

 17   consideration under the animal rule.  We have had

 

 18   sponsors come in and propose to pursue a drug under

 

 19   the animal rule where there was no activity data in

 

 20   clinical trials in adults, let's say, as applied to

 

 21   what we are considering today.  The important thing

 

 22   to consider is in what way can this principle be

 

 23   applied to pediatric oncology drugs.  I think this

 

 24   is something that maybe would be worth discussing.

 

 25             Juvenile animal studies can be useful for

 

                                                               277

 

  1   safety determinations.  They are not prohibitively

 

  2   challenging to conduct.  You can dose rat pups from

 

  3   day seven on.  In fact, people have even looked at

 

  4   beginning with birth, transferring drug in mother's

 

  5   milk and then starting to dose after weaning.

 

  6   There are all kinds of ways you can manipulate

 

  7   neonatal animal studies.

 

  8             The available data doesn't indicate that

 

  9   juvenile animal studies need to be routinely

 

 10   conducted, but they might be needed under certain

 

 11   circumstances, as I have mentioned previously.  But

 

 12   the database is limited and this conclusion could

 

 13   change.  I don't think it will but, as with

 

 14   anything, as we start seeing more juvenile animal

 

 15   studies we will start looking back at these and

 

 16   deciding whether or not we made the right decision

 

 17   in our recommendation.

 

 18             So, thanks and I appreciate your

 

 19   attention.

 

 20             DR. SANTANA:  Thank you.  I am going to

 

 21   take the chair's prerogative and ask you two

 

 22   questions because I don't want you to leave the

 

 23   podium without addressing these.  One is, can you

 

 24   give us an idea of the universe of where this is

 

 25   applied?  I mean, how many times when there is a

 

                                                               278

 

  1   new drug, either in development or a drug that is

 

  2   already out there, are we going back and doing

 

  3   either retrospectively, when is the drug is already

 

  4   out there or as part of the development plan, some

 

  5   of these studies addressing specific issues of

 

  6   toxicity?  Is this a common thing that happens?

 

  7             DR. HASTINGS:  In juvenile animals?

 

  8             DR. SANTANA:  Yes, is this common or

 

  9   uncommon?  That is the first question.  Then a

 

 10   corollary to that is, are there specific animal

 

 11   models that address specific systems?  So, is there

 

 12   an animal model that already looks at neurologic

 

 13   toxicity?  Is there an animal model that already

 

 14   looks at cardiac?  Or, is it really just this model

 

 15   and then we look the nervous system or we look for

 

 16   the heart system, and so on and so forth?

 

 17             DR. HASTINGS:  Well, the first question,

 

 18   yes, we have seen a number of juvenile animal

 

 19   studies.  Dr. Karen Davis Bruno, who is the chair

 

 20   of that committee, has been keeping a running

 

 21   tabulation.  Karen, do you know what the number is

 

 22   right now?

 

 23             DR. BRUNO:  [Not at microphone; inaudible]

 

 24             DR. HASTINGS:  Also, as I understand it

 

 25   from sponsors, when it was understood that we were

 

                                                               279

 

  1   working on this guidance, if they were going to

 

  2   pursue pediatric development before they understood

 

  3   that we were looking at developing a for-cause

 

  4   guidance, in fact, a number of sponsors just did

 

  5   them.  I mean, they basically just said we are

 

  6   going to anticipate that FDA is going to ask for

 

  7   them.  So, yes, there are a number of them and some

 

  8   of them have been quite informative.  I didn't

 

  9   really get into that because, frankly, I am not

 

 10   aware of a case in pediatric oncology.

 

 11             As far as a preferred animal, well, no.  I

 

 12   wish we could say that there is.  You know, we have

 

 13   standard models in toxicology in drug

 

 14   development--rats, beagle dogs, cynomolgus monkeys

 

 15   and it is almost like those are the better models

 

 16   simply because we have just developed so much data

 

 17   with them that we understand what is going on

 

 18   there.  If it is neurological though, you are

 

 19   probably wanting to think more in the line of a

 

 20   non-human primate like cynomolgus.  But for, like,

 

 21   immune parameters probably rats would be a better

 

 22   model simply because we have the reagents to do

 

 23   that kind of study.

 

 24             DR. SANTANA:  Thank you for answering

 

 25   those two questions.  I think they were relevant to

 

                                                               280

 

  1   what you were trying to address in your

 

  2   presentation.  I will invite Malcolm--he has the

 

  3   daunting task of being the last speaker.

 

  4           Assessing Anti-Tumor Activity in Nonclinical

 

  5                    Models of Childhood Cancer

 

  6             DR. SMITH:  I would like to thank Steve

 

  7   and colleagues at the FDA for sponsoring this

 

  8   meeting and for the invitation to speak here this

 

  9   afternoon.

 

 10             I will be talking about NCI's initiatives

 

 11   to develop nonclinical models for pediatric

 

 12   oncology.  Throughout the talk I will slip between

 

 13   nonclinical and preclinical.  The slides are

 

 14   variably labeled that way but you will know what I

 

 15   mean.  The three major things I will be focusing on

 

 16   are, one, why we need to be working in this area;

 

 17   two, why we are doing what we are doing; and,

 

 18   three, why we think it has at least some chance of

 

 19   providing useful information.

 

 20             I have shown this slide at I think

 

 21   previous pediatric ODAC meetings, but it is the

 

 22   drug development pyramid and it makes the point

 

 23   that there are more agents entering Phase I studies

 

 24   in adults than we can move into children; then,

 

 25   more agents during Phase I in children than we can

 

                                                               281

 

  1   conduct Phase II studies for; then only a very

 

  2   limited number of Phase III studies that we can

 

  3   conduct.  We are not limited now at the Phase I

 

  4   setting.  We actually could study more drugs in the

 

  5   Phase I setting.  Where we really are limited is in

 

  6   moving to Phase II and doing all the Phase II and

 

  7   pilot studies that we need with these new agents,

 

  8   and then especially moving into Phase III studies

 

  9   and the one neuroblastoma Phase III study or

 

 10   rhabdomyosarcoma Phase III study that we may be

 

 11   able to do in the next three, four to five years.

 

 12             To make a concrete example of this

 

 13   neuroblastoma and looking at the agents under

 

 14   evaluation now, these are all in pediatric Phase I

 

 15   or Phase II trials--a demethylating agent,

 

 16   decitabine, fenretinide, interleukin-12, the Trk

 

 17   tyrosine kinase inhibitor, oxaliplatin, HDAC

 

 18   inhibitors and then BSO.  Those are the single

 

 19   agents or we could combine those with standard

 

 20   chemotherapy agents in different regimens.  We can

 

 21   combine them with each other and try to inhibit

 

 22   some of the different pathways jointly that these

 

 23   agents inhibit.  So, how are we going to pick which

 

 24   of these agents, which combinations to bring

 

 25   forward for the one neuroblastoma Phase III study

 

                                                               282

 

  1   that we will be starting in two or three years?  It

 

  2   is a daunting challenge to try to get data that

 

  3   informs that decision.

 

  4             Hence, this is a primary need for some

 

  5   help with that from the preclinical or nonclinical

 

  6   area.  If we had predictive nonclinical methods, it

 

  7   could contribute to prioritizing agents for

 

  8   evaluation against specific types of childhood

 

  9   cancer.  To do this, we need a systematic approach

 

 10   opposed to what really has been a haphazard

 

 11   approach over the past twenty years.  The

 

 12   systematic approach is required to assess the

 

 13   predictive value of pediatric nonclinical models.

 

 14             In recognition of the need for such a

 

 15   systematic approach, the NCI board of scientific

 

 16   advisors approved committing ten million dollars to

 

 17   this effort over the next five years through the

 

 18   Pediatric Preclinical Testing Program.  I will

 

 19   describe this in a bit more detail later but for

 

 20   now suffice it to say that this will be a

 

 21   systematic approach, primarily based on in vivo

 

 22   testing with xenograft models, but also having an

 

 23   in vitro component and making use of genetically

 

 24   engineered models when those are available and

 

 25   applicable.

 

                                                               283

 

  1             So, the questions I am asked about this

 

  2   when I have talked about this are, well, why are

 

  3   you doing this?  Don't you know that adults have

 

  4   used xenografts and xenografts don't

 

  5   work?--analogous to Pat Reynolds' question earlier.

 

  6   I would respond to this by pointing out three

 

  7   papers, and I will start with the last one, a

 

  8   review article that I would refer you to for

 

  9   marshaling of the arguments that xenografts can

 

 10   contribute to drug development and the take-home

 

 11   message there is better than commonly perceived but

 

 12   can be improved.

 

 13             The first reference was a paper from the

 

 14   developmental therapeutics program at NCI, and the

 

 15   conclusion there was that although maybe a breast

 

 16   cancer xenograft didn't predict for activity in

 

 17   breast cancer, activity across a range of

 

 18   xenografts predicted that that was an agent that

 

 19   had a good chance of being successful when

 

 20   transferred to the clinic, not necessarily for the

 

 21   tumors that weren't in the xenograft models but for

 

 22   at least some cancers having activity.

 

 23             The second paper, a more recent paper

 

 24   published last year in Clinical Cancer Research,

 

 25   made the point that using panels of xenografts for

 

                                                               284

 

  1   a given tumor type increases the likelihood for

 

  2   correct prediction, and we will be focusing on

 

  3   panels of xenografts in our preclinical testing

 

  4   program.

 

  5             This shows two figures from that paper.

 

  6   If you look at the one on your left, each of the

 

  7   squares represents a drug that was studied in the

 

  8   clinic.  There is the Phase II activity, the

 

  9   response rate.  And, it was studied in a panel of

 

 10   xenografts, and the readout there is the mean

 

 11   treatment to control.  So, a low treatment to

 

 12   control indicates a high level of activity in the

 

 13   preclinical setting and high response rate in the

 

 14   Phase II, of course, indicates high activity there.

 

 15   So, you see the predictive value for at least two

 

 16   of these xenograft panels where activity in the

 

 17   preclinical setting in these ovarian xenograft

 

 18   panels and the non-small lung cancer xenograft

 

 19   panels predicted for Phase II activity for these

 

 20   agents.

 

 21             The other point that I make when

 

 22   justifying why we think this has some reasonable

 

 23   chance of being successful is that we have the

 

 24   advantage of being able to make use of pharmacology

 

 25   to enhance a predictive ability of preclinical

 

                                                               285

 

  1   models.  We will be able to make comparisons

 

  2   between mouse pharmacology to human pharmacology

 

  3   and this can rule out the trivial explanation for

 

  4   activity in xenograft models.  That trivial

 

  5   explanation for an agent being active in a

 

  6   xenograft model being that the mice tolerate much

 

  7   more of the agent than humans do.  So, a human

 

  8   cancer implanted in the mice is going to be exposed

 

  9   to much higher levels than we will ever seen in the

 

 10   clinical setting and there is a good chance that

 

 11   activity will be seen but it won't be replicated in

 

 12   humans.

 

 13             In the pediatric preclinical setting we

 

 14   can use both the activity of the agent in our

 

 15   pediatric preclinical models that test results, and

 

 16   also the comparison of the mouse PK of the agent

 

 17   with the PK of the agent in the initial adult

 

 18   trials.  We will be studying these agents or we

 

 19   will be making our decision at a time after we have

 

 20   some initial adult experience.

 

 21             So, the most promising agents then will be

 

 22   those that have activity in the pediatric models at

 

 23   serum levels that are actually achievable or

 

 24   systemic exposures that are achievable in humans.

 

 25   Peter Houghton gave examples of this and I will

 

                                                               286

 

  1   just reiterate two of those.  The topo-1

 

  2   inhibitors, irinotecan where incorporating PK led

 

  3   to positive prediction for the activity of

 

  4   irinotecan against neuroblastoma.  Then,

 

  5   incorporating PK correctly predicted inactivity for

 

  6   another agent that he described, sulofenur.

 

  7             Peter mentioned the data that we have that

 

  8   support the potential for prediction, and I just

 

  9   list those, the data that he described for activity

 

 10   of agents in rhabdomyosarcoma xenografts mirroring

 

 11   the clinical activity of these agents, the correct

 

 12   prediction of activity for topo-1 agents against

 

 13   both rhabdomyosarcoma and neuroblastoma.  Another

 

 14   point is that models now are not just limited to

 

 15   rhabdomyosarcoma and neuroblastoma.  Peter

 

 16   described the Wilms tumor and some of the

 

 17   predictive supportive data there.

 

 18             Importantly, we also have xenografts for

 

 19   acute lymphoblastic leukemia.  Since this is the

 

 20   most common cancer in children and a major cause of

 

 21   mortality among children with cancer, it will be

 

 22   important to also look at this in an in vivo

 

 23   preclinical setting.

 

 24             This is work from Richard Lock, published

 

 25   in Blood a couple of years ago, just showing the

 

                                                               287

 

  1   blast cells in the patient and then growing in the

 

  2   NOD/SCID mice.

 

  3             This is a table from that work showing

 

  4   that when these lines are transplanted into mice

 

  5   with no treatment there is a reasonably consistent

 

  6   growth pattern.  With treatment with an agent known

 

  7   to be active against some childhood ALL cases there

 

  8   is substantial growth delay for some cases;

 

  9   moderate growth delay for other cases; and no

 

 10   growth delay for some.   Importantly, this in vivo

 

 11   sensitivity to vincristine correlated with what we

 

 12   know is an important measure of sensitivity in ALL,

 

 13   the duration of the first complete remission.  So,

 

 14   we have the capability now to look at these ALL

 

 15   xenografts to address this important disease.

 

 16             An important contribution of the

 

 17   preclinical models now is in the area of

 

 18   molecularly targeted agents, and the ability to

 

 19   make preclinical pharmacokinetic and

 

 20   pharmacodynamic comparisons.  Peter mentioned this

 

 21   and I will reiterate it.  Especially important in

 

 22   this era of molecular targets, we can use these

 

 23   models to identify the degree of target modulation

 

 24   that is associated with anti-tumor activity, 50

 

 25   percent inhibition, 75 percent, 90 percent, what is

 

                                                               288

 

  1   needed in order to achieve anti-tumor activity; how

 

  2   long does target modulation need to occur to

 

  3   achieve the desired effect; and then particularly

 

  4   important for children, what are the serum levels

 

  5   or systemic exposures of the agent that are

 

  6   associated with the requisite levels of target

 

  7   modulation because it is going to be very difficult

 

  8   for most childhood solid tumors especially to be

 

  9   able to biopsy repeatedly tumor specimens to

 

 10   measure this in children so we can understand the

 

 11   pharmacology in children and target the systemic

 

 12   levels that we have shown in the preclinical models

 

 13   to achieve the desired level of target modulation.

 

 14   This is also an opportunity to correlate anti-tumor

 

 15   activity with gene expression profiles and protein

 

 16   expression profiles.

 

 17             One area that we are working in to try to

 

 18   facilitate the evaluation of molecular targeted

 

 19   agents is a project called POPP-TAP, or the

 

 20   Pediatric Oncology Preclinical Protein and Tissue

 

 21   Array Project.  This is a collaboration between

 

 22   NCI, both intramural and extramural, and Children's

 

 23   Oncology Group researchers.  The objective of this

 

 24   collaboration is to develop tissue and cell arrays

 

 25   and protein lysate arrays of pediatric preclinical

 

                                                               289

 

  1   cancer models, primarily focusing initially on

 

  2   xenografts and we are going to have close to 100

 

  3   xenografts, different xenografts for which we will

 

  4   have these tissue arrays available for study by

 

  5   researchers.  Also, Kahn's laboratory is

 

  6   determining the gene expression profiles for these

 

  7   pediatric preclinical cancer models, again focusing

 

  8   initially on almost 100 xenografts for this.  Then,

 

  9   these data will be available for researchers as

 

 10   well.  We hope that this project will facilitate

 

 11   the conduct and interpretation of preclinical

 

 12   testing of targeted agents in childhood cancer

 

 13   models.

 

 14             The kind of complicating factors in

 

 15   testing molecularly targeted agents--the comment is

 

 16   sometimes made, well, you know the target is there,

 

 17   just go after the tumors that express the target.

 

 18   It is not that easy.  One of the complicating

 

 19   factors is the promiscuity of agents.  A targeted

 

 20   agent may hit multiple targets, some recognized;

 

 21   some not.  The Bay compound is one of many

 

 22   examples.  It was initially a raf kinase inhibitor.

 

 23   So, there is promiscuity of agents in terms of

 

 24   their targets.

 

 25             There are multiple biological effects of

 

                                                               290

 

  1   modulating a particular target of these so-called

 

  2   molecularly targeted agents so farnesyl transferase

 

  3   inhibitor in all the pathways that affects; the

 

  4   proteasome inhibitors in all the different pathways

 

  5   that that affects; Hsp90 inhibitors, all the

 

  6   pathways affected there.  And, it is very hard,

 

  7   kind of on first principles of tumor biology, to

 

  8   predict a priori what the potential applicability

 

  9   of a particular agent such as this is to a

 

 10   particular childhood cancer based on just its

 

 11   biology.  The preclinical testing then can allow

 

 12   identification of previously unrecognized or

 

 13   unsuspected activities that may have clinical

 

 14   relevance.

 

 15             I am often asked, in terms of addressing

 

 16   preclinical activities, well, what about mouse

 

 17   genetic models?  Why aren't you focusing solely on

 

 18   mouse genetic models?  They have certainly made

 

 19   critical contributions to our understanding of

 

 20   cancer pathogenesis.  In order to use genetic

 

 21   models for testing, not all models will be

 

 22   appropriate for testing.  Really specific

 

 23   properties are needed, particularly short latency

 

 24   and high penetration for feasible testing are two

 

 25   characteristics needed and not all models have

 

                                                               291

 

  1   that.

 

  2             But there are some genetically engineered

 

  3   models for pediatric cancers that may have these

 

  4   characteristics and be suitable for drug testing.

 

  5   For example, the MYCN model for neuroblastoma may

 

  6   be appropriate and we will try to use that if we

 

  7   can.

 

  8             The other caution is that a mouse is a

 

  9   mouse, and mouse biology is not the same as human

 

 10   biology.  So, the lessons from the mouse genetic

 

 11   models may not apply directly to the human setting.

 

 12   There was an excellent review last year that really

 

 13   documented this issue and made the point that more

 

 14   humanized mice may more faithfully replicate human

 

 15   cancers.

 

 16             The preclinical testing program that we

 

 17   have worked on over the last year or two to

 

 18   initiate will be based on panels of xenograft lines

 

 19   for the most common childhood cancers.  It will

 

 20   incorporate an in vitro testing component along the

 

 21   lines that Peter Adamson outlined, particularly in

 

 22   areas like the combination studies which may

 

 23   provide valuable information.

 

 24             We hope to be able to systematically test

 

 25   10-15 agents per year, seeking to obtain agents

 

                                                               292

 

  1   near the time that a commitment is made for the

 

  2   initial evaluation in adults so that, by the time

 

  3   the adult clinical experience is available and

 

  4   there is evidence that this may be an agent that

 

  5   could be studied in children, we will have

 

  6   preclinical data to better address the question of

 

  7   whether this is an agent that should be studied in

 

  8   children.  This will be implemented via a contract

 

  9   mechanism with the primary contractor and the

 

 10   potential for subcontracts for testing specific

 

 11   cancer types.

 

 12             The schema that Peter showed is shown

 

 13   here.  I will just make the point here that we will

 

 14   be using panels of tumors.  For example, if this is

 

 15   a rhabdomyosarcoma, each panel is represented by 6

 

 16   to 8 to 10 different xenografts, and then testing

 

 17   at the MTD initially.  When hits are identified,

 

 18   activity is identified, then being able to go and

 

 19   study the agent more intensively, look at a full

 

 20   dose response, obtain PK data if that is not

 

 21   already available, and do some of the molecular

 

 22   studies if those are warranted.

 

 23             A critical issue is addressing the

 

 24   intellectual property issues.  We have made efforts

 

 25   over the past years to develop, in collaboration

 

                                                               293

 

  1   with academic investigators and pharmaceutical

 

  2   sponsors, a model MTA.  This model MTA will be used

 

  3   for all transfers by companies of their proprietary

 

  4   compounds to NCI-supported investigators for

 

  5   preclinical testing.  Acceptance of the model MTA,

 

  6   and it was included in the RFP for establishing the

 

  7   preclinical testing program, but acceptance of the

 

  8   model MTA is a requirement for participation in the

 

  9   program.

 

 10             I actually have some copies of the model

 

 11   MTAs.  There is one for transfer of the agent to

 

 12   MCI and there is one for transfer of the agent from

 

 13   MCI to the test sites.  But Dr. Sherry Ansher is

 

 14   the CTEP contact for those.  If anyone wanted

 

 15   copies, I would be glad to provide those to you.

 

 16             In summary and in closing, appropriate

 

 17   prioritization is key to future treatment advances

 

 18   for childhood cancer.  If we make good decisions in

 

 19   terms of which agents we bring forward, and

 

 20   particularly to the Phase III setting, we have a

 

 21   chance for making advances.  if we don't, then our

 

 22   advances will be limited.

 

 23             The Pediatric Preclinical Testing Program

 

 24   may contribute to successful prioritization but

 

 25   systematic preclinical testing of all agents

 

                                                               294

 

  1   entering clinical evaluation in children should

 

  2   become the standard of care, not because we know

 

  3   what to do with these data now--we may have ideas

 

  4   of what to do with these data, but because a

 

  5   systematic approach is what we need to allow

 

  6   validation of the panels and to optimize the

 

  7   pediatric preclinical tumor panels.  Thank you and,

 

  8   again, thanks to the FDA for this opportunity.

 

  9                       Committee Discussion

 

 10             DR. SANTANA:  Thank you, Malcolm.  We have

 

 11   a few minutes for questions for presenters before

 

 12   we go into the period of answering the questions.

 

 13   Dr. Przepiorka?

 

 14             DR. PRZEPIORKA:  Thanks.  Two questions,

 

 15   one for either Malcolm or Peter.  Peter had a slide

 

 16   up there of I think it was MMI114 looking at a

 

 17   single dose or dose schedule against a series of

 

 18   tumors.  If I recall, your conclusion was it was

 

 19   not a very active drug because the AUC was ten

 

 20   times greater than what one could expect to achieve

 

 21   in humans.  I was somewhat disappointed because I

 

 22   could think of three or four drugs that we already

 

 23   use for which we could probably have made the same

 

 24   conclusion based on a single dose schedule being

 

 25   tested.

 

                                                               295

 

  1             So, my question for either of you is,

 

  2   especially with the development of the new program,

 

  3   is there an established panel of dose schedules

 

  4   that will be used for drug testing so that you know

 

  5   when a single high dose is going to be effective as

 

  6   opposed to low continuous exposure before a drug is

 

  7   thrown out?

 

  8             DR. HOUGHTON:  I think in the case of

 

  9   NGI114 we have basically done other schedules.  I

 

 10   think what we would hope is that a fair amount of

 

 11   optimization will have been done if we get a drug

 

 12   from industry that is going into a clinical trial,

 

 13   that a lot of the various schedules that have been

 

 14   examined and information on which are the best

 

 15   schedules will be made available at that point.  I

 

 16   think if you look at the size of the screening

 

 17   program, if we went to doing the classic schedules

 

 18   that you are going to use in the clinic, I don't

 

 19   think the screening program has the capacity for

 

 20   those; it certainly doesn't have the funding to do

 

 21   that.  So, I think for most drugs that will come

 

 22   from industry, they may well have that information

 

 23   already so that would at least allow us to do the

 

 24   first cut using the optimal schedule that they have

 

 25   and, in most cases, those have been quite accurate.

 

                                                               296

 

  1             DR. PRZEPIORKA:  If one has knowledge of

 

  2   the mechanism of action and the pharmacokinetics,

 

  3   could one potentially come up with the three best

 

  4   guesses and so not have to do a whole bunch of

 

  5   different dose schedules, or is that not a

 

  6   reasonable approach?

 

  7             DR. HOUGHTON:  Again, a lot of that

 

  8   information will be available to guide how we test

 

  9   the drug in the screening program.  I think we

 

 10   still have to go to the MTD.  I think that is

 

 11   probably appropriate because one of the things you

 

 12   want to do is get some idea of the tumor

 

 13   sensitivity relative to an MTD in the mouse so

 

 14   ultimately you want to do that with respect to

 

 15   pharmacokinetics.  So, irrespective of whether you

 

 16   know the mechanism of the action of the drug, I

 

 17   think the consensus was that you go for the MTD

 

 18   even if you have a molecularly targeted drug where

 

 19   you think it is a specific kinase inhibitor.  That

 

 20   is for two reasons.  One is if you see no activity

 

 21   that probably tells you that, you know, this is not

 

 22   a drug that is suitable for treating certain

 

 23   pediatric cancers.  The other is that despite

 

 24   having very strong evidence that a specific target

 

 25   is, indeed, the target, when you go to the MTD you

 

                                                               297

 

  1   may, in fact, reveal additional activities.  I

 

  2   think what we are trying to do is the minimum

 

  3   amount of work, not because we don't like to do any

 

  4   work but the minimum amount of work means minimum

 

  5   utilization of resources to do a first cut to

 

  6   identify those drugs that are worth pursuing, and

 

  7   maybe looking at scheduling issues but to eliminate

 

  8   those where we feel there is very little reason to

 

  9   pursue that.

 

 10             DR. PRZEPIORKA:  If Dr. Chand Khanna is

 

 11   still here, I have a question.  I mean, GLP came

 

 12   around because of some major issues in drug

 

 13   development for adults and I would hate to see the

 

 14   same problems arise in pediatrics because GLP was

 

 15   not applied.  The comment was made earlier that it

 

 16   is too expensive for a vivarian in academia to

 

 17   actually run under GLP but, given all the rules

 

 18   that govern how you deal with animal care nowadays,

 

 19   I can't imagine that it is not already running

 

 20   under GLP.  Does the Center for Comparative

 

 21   Oncology animal housing at NCI--in your experience,

 

 22   is that run under GLP and is it really a stretch to

 

 23   try to get everybody who is going to be doing

 

 24   preclinical testing to do that?

 

 25             DR. SANTANA:  Can you come to the

 

                                                               298

 

  1   microphone, please?

 

  2             DR. HIRSCHFELD:  While he is coming, I

 

  3   want to request the permission of the chair to have

 

  4   Dr. Khanna and Dr. Meltzer take some empty seats at

 

  5   the table and to have them join in the discussion.

 

  6   I think there are empty seats between Dr. Weiner

 

  7   and Ms. Haylock and there is an empty seat next to

 

  8   Ms. Ettinger.

 

  9             DR. KHANNA:  Yes, a point of

 

 10   clarification, at NCI we actually aren't going to

 

 11   be managing pet animals in trials.  We will be

 

 12   managing those trials through veterinary teaching

 

 13   hospitals that do operate under GCP guidelines in

 

 14   many situations.  So, that GCP hurdle is certainly

 

 15   passed at many of those sites that we will be

 

 16   working with and, in fact, it will be a requirement

 

 17   for them to be involved in our cooperative groups.

 

 18             DR. SANTANA:  Donna, not to take this

 

 19   discussion down a different route, but those for us

 

 20   who are not familiar with the issues related to

 

 21   GLP, since you hinted that there was an issue,

 

 22   could somebody summarize what those are?

 

 23             DR. HIRSCHFELD:  I think we have a lot of

 

 24   experts in the room but, in brief, the

 

 25   International Conference on Harmonization, as well

 

                                                               299

 

  1   as the FDA, have adopted standards under which

 

  2   animal studies are conducted.  These standards

 

  3   collectively are referred to as Good Laboratory

 

  4   Practice.  Is Dr. Hastings still here?  Do you want

 

  5   to add anything to that?

 

  6             DR. HASTINGS:  [Not at microphone;

 

  7   inaudible]

 

  8             DR. SANTANA:  Please us the microphone

 

  9   because we really need to listen to the discussion

 

 10   and sometimes it is difficult, and also record it

 

 11   for the record.  You can take the podium; that

 

 12   would be fine.

 

 13             FDA PARTICIPANT:  GLP has many components

 

 14   to it.  It includes the test article and how stable

 

 15   it is.  The composition has to do with the people

 

 16   that are involved with the research, like their CV

 

 17   being on line, how they have been trained.  It has

 

 18   to do with the instrumentation and how they are

 

 19   calibrated or if they are appropriate for the

 

 20   testing that is being done.  It has to do with the

 

 21   animal husbandry, and how they are kept, the room

 

 22   and the building, and many other components to it.

 

 23             DR. SANTANA:  Donna, did you want to

 

 24   elaborate on that?

 

 25             DR. PRZEPIORKA:  Yes, I think from all the

 

                                                               300

 

  1   talks that I have sat through, all the way back to

 

  2   orientation, I believe GLP came around as a result

 

  3   of some issues regarding fraud and poor science in

 

  4   the late '60s, early '70s.  I was just looking to

 

  5   see if the poster was still up because I think I

 

  6   remember the poster being up during orientation.

 

  7   So, GLP came around as a result of a lot of

 

  8   problems with scientific integrity in the initial

 

  9   preclinical work that was handed in with drug

 

 10   trials supporting FDA approval, and to have that

 

 11   happen in the pediatric setting right now would

 

 12   probably be a huge step backwards for pediatric

 

 13   drug development.

 

 14             DR. ADAMSON:  I just want to clarify that

 

 15   there is a difference between GLP and GCP.  Without

 

 16   question, pediatric trials are according to GCP.

 

 17   What you are saying is the animal clinical trials

 

 18   are going to be conducted according to GCP.  That

 

 19   is a different level of work but that is the

 

 20   standard in pediatric drug development trials.

 

 21             GLP, as we have just heard--there are very

 

 22   few academic laboratories, adult or pediatric, that

 

 23   do work according to GLP.  That is the reality of

 

 24   academic laboratories.  There are very few that do

 

 25   it according to GLP because the costs become

 

                                                               301

 

  1   prohibitive.  There are some laboratories that can

 

  2   do it but I think they are a distinct minority.

 

  3   Without question, would every place like to do it

 

  4   according to GLP?  Yes, but the funding is simply

 

  5   not there to meet those costs.

 

  6             DR. WILLIAMS:  I must say that working

 

  7   with our pharm tox colleagues we do not demand GLP

 

  8   when we see a new IND, but we do demand that they

 

  9   analyze where it differs from GLP and justify those

 

 10   differences.

 

 11             DR. SANTANA:  Malcolm, were you going to

 

 12   make an additional comment?

 

 13             DR. SMITH:  In the RFP for the preclinical

 

 14   contract we did not specify GLP.  That was at the

 

 15   recommendation of colleagues in the Developmental

 

 16   Therapeutics Program.  You know, basically it is

 

 17   what Peter was saying, that it would limit the pool

 

 18   of researchers who could do that work.  We will

 

 19   have appropriate procedures in place so the

 

 20   credibility of the results will, we hope, be above

 

 21   question but we have not required that they meet

 

 22   the GLP requirements in the RFP.

 

 23             DR. SANTANA:  Dr. Hirschfeld?

 

 24             DR. HIRSCHFELD:  I will make one further

 

 25   comment and then maybe we can go to the questions. 

 

                                                               302

 

  1   GLP I think is more precise than GCP.  GCP is very

 

  2   open to interpretation and that was one of the

 

  3   rationales for having our discussion this morning,

 

  4   and it is a continuing source of guidances and

 

  5   directives and other documents attempting to decide

 

  6   how GCP can be applied to any particular study,

 

  7   whereas GLP tends to be more explicit.

 

  8             DR. SANTANA:  Good.  Any other questions

 

  9   to the panel members or discussants?

 

 10             DR. REYNOLDS:  I wanted to tie a little

 

 11   bit of what Eric was saying this morning to

 

 12   comments made by Peter and particularly by Malcolm

 

 13   where you suggested a standard of care would be

 

 14   preclinical testing if we are engaging in human

 

 15   studies in pediatrics.  I think it would seem that

 

 16   given what Eric was saying--this was really not a

 

 17   point of discussion in the morning when we were

 

 18   talking about monitoring but he did point out the

 

 19   sort of ethics dilemma involved in facing a Phase I

 

 20   study where you are looking at having to deliver

 

 21   some prospect of benefit to a patient in the

 

 22   context of doing the study.  I think I would like

 

 23   to suggest that we incorporate or think about some

 

 24   sort of way that the agency might incorporate

 

 25   Malcolm's suggestion of a standard of care, of

 

                                                               303

 

  1   having some sort of preclinical data in the

 

  2   pediatric tumor setting before engaging in testing

 

  3   these agents in the pediatric setting.

 

  4             DR. SANTANA:  Peter?

 

  5             DR. ADAMSON:  I would actually put out a

 

  6   caveat that that would be a goal to try to realize

 

  7   perhaps within the next five or ten years.  The

 

  8   large majority of agents today that are active

 

  9   drugs for children with cancer have not gone

 

 10   through preclinical testing.  There is a lot of

 

 11   inactivity in industry with very important drugs.

 

 12   So, it is an ideal we would like to move towards

 

 13   but I think we are many steps away before saying

 

 14   that that is the standard of care.

 

 15             DR. SANTANA:  Dr. Smith?

 

 16             DR. SMITH:  The other caveat would be that

 

 17   in the future it could be a standard of care

 

 18   because we have predictive models that we are

 

 19   confident of and it makes sense to act on our

 

 20   knowledge of these predictive models.  That is in

 

 21   the future, why it should be the standard of care.

 

 22   Why it should be the standard of care now is

 

 23   because if we don't do it systematically and obtain

 

 24   the experience, then we won't ever get to that

 

 25   future.  So, for now the standard of care is

 

                                                               304

 

  1   because only by systematically approaching this

 

  2   problem can we develop the data that gets us to the

 

  3   point where we are confident making decisions based

 

  4   on these data.

 

  5             DR. REYNOLDS:  Absolutely, but I think

 

  6   what Peter said in one of his slides is quite true,

 

  7   and that is under the ideal circumstances of a good

 

  8   laboratory model, if you can't get good responses

 

  9   in your disease type you are probably unlikely,

 

 10   almost assuredly unlikely to get those responses in

 

 11   the children.  So, I think that doing some testing

 

 12   to exclude agents that we then would not be

 

 13   exposing children to when they have no prospect of

 

 14   benefit based upon what is probably a predictive

 

 15   model--that is, if you don't get any activity in

 

 16   the lab you are probably not going to get it in the

 

 17   clinic--should be at least a consideration.

 

 18             DR. SANTANA:  Dr. Houghton, I think you

 

 19   had a comment?

 

 20             DR. HOUGHTON:  Only to add that I think in

 

 21   five years time we will have a much better idea of

 

 22   whether this is correct or not.  I think the one

 

 23   thing that perhaps didn't come out strongly enough

 

 24   from maybe the three of us is that what we are

 

 25   proposing in terms of PPTP is an experiment and we

 

                                                               305

 

  1   don't know how accurate the models are going to be.

 

  2   We don't know what the flaws or the limitations

 

  3   are.

 

  4             So, in a way, I think it would be also

 

  5   inappropriate if you had no activity in the model

 

  6   not to pursue that at a clinical level because, in

 

  7   fact, they may be very important experiments that

 

  8   will reveal the fact that the models have

 

  9   limitations.  What we want to know at the end of

 

 10   the day is with we are on the right track or the

 

 11   wrong track, and if there are limitations to try

 

 12   and address those in the next generation of models.

 

 13             One of the biggest problems I see in the

 

 14   development of models in preclinical development,

 

 15   in the thirty years I have been playing this game,

 

 16   is that we have these transitions, we transitioned

 

 17   from syngeneic rodent models to xenografts, to in

 

 18   vitro systems to xenografts, to perhaps transgenics

 

 19   and nobody has taken the time to look back to see

 

 20   what the problems were of the previous model that

 

 21   would then allow us to develop a better model.  So,

 

 22   the next five years may be very revealing in terms

 

 23   of the current models we have and their limitations

 

 24   but give us the information that the next

 

 25   generation of models won't make the same mistakes

 

                                                               306

 

  1   as the previous models.

 

  2                     Questions for Discussion

 

  3             DR. SANTANA:  With those words of wisdom

 

  4   and advice to all of us, let's go ahead and try to

 

  5   tackle the questions for discussion.  FDA is

 

  6   requesting that we comment on three issues and, for

 

  7   the record, I will go ahead and read the

 

  8   introduction and the questions.

 

  9             Because of the limited number of pediatric

 

 10   oncology patients and because of the problems

 

 11   unique to pediatric drug development, it may not

 

 12   always be feasible to evaluate all aspects of

 

 13   efficacy and safety in clinical studies.  In some

 

 14   settings, extrapolation of results from nonclinical

 

 15   studies may be appropriate.

 

 16             The first question is what types of

 

 17   questions that are of potential clinical relevance

 

 18   but are not feasible or acceptable to answer in a

 

 19   clinical study could be addressed by nonclinical

 

 20   studies?  Then various examples are given after the

 

 21   question that potentially could fit the answer that

 

 22   we are being asked to provide.

 

 23             I want to comment that one of the things

 

 24   that I gathered from some of the discussion and

 

 25   presentation this afternoon is that some of the

 

                                                               307

 

  1   field is moving to molecularly targeted therapies,

 

  2   whatever that means, and we may have limitations in

 

  3   our patients in being able to correctly or early on

 

  4   assess the correct target or do multiple biopsy

 

  5   samples, etc., to see whether relevant targets are

 

  6   being affected.  I think in that setting, in which

 

  7   the ethical issue of providing multiple biopsies in

 

  8   a patient may be relevant or may not make the

 

  9   clinical studies feasible, these models could be

 

 10   used to address those very early on so that when we

 

 11   get to the stage of testing these drugs in

 

 12   patients, then sampling strategy may be very

 

 13   limited or may be focused to such a degree that

 

 14   ethically it doesn't become a constraint for the

 

 15   study.  So, that is one setting where I think some

 

 16   of these preclinical models potentially could help

 

 17   us in terms of limiting the ethical barriers we may

 

 18   have when we introduce these molecularly targeted

 

 19   drugs to our trials.  That is one example that I

 

 20   think would be relevant.  Dr. Reynolds?

 

 21             DR. REYNOLDS:  I would like to suggest

 

 22   another example.  If one is dealing with agents,

 

 23   two new molecular entities or new agents of which

 

 24   one may have some modest activity and the other, as

 

 25   a single agent, may have very little activity but

 

                                                               308

 

  1   in combination in preclinical studies have striking

 

  2   synergy, requiring that you demonstrate activity

 

  3   for each individual agent in a patient, whereas if

 

  4   you went in with the combination you might get

 

  5   striking activity, and using the preclinical data

 

  6   or nonclinical data, however you want to describe

 

  7   it, to justify the approval of the agent as a

 

  8   combination I think would make some sense, and

 

  9   would spare children the ethical dilemma of being

 

 10   treated potentially with an agent that is predicted

 

 11   by preclinical data to be fairly non-effective, yet

 

 12   might contribute to the overall response of the two

 

 13   agents in combination.

 

 14             DR. SANTANA:  Dr. Adamson?

 

 15             DR. ADAMSON:  In looking at the examples,

 

 16   Steve, that you have here, almost all of them are

 

 17   looking at host and not tumor.  I think that is

 

 18   fine and helps us think about what you are after.

 

 19   What I would caution is that we don't know, even as

 

 20   far as host response or, you know, developing

 

 21   animal models, how predictive they really are, and

 

 22   the experience with the fluoroquinolones I think is

 

 23   a good one.  We are using them and we are still

 

 24   learning what the real risk is.  We should not

 

 25   delay the initiation of pediatric testing of

 

                                                               309

 

  1   anti-cancer agents for the results of these types

 

  2   of studies because in the balance, of course, are

 

  3   diseases that carry a far more certain outcome for

 

  4   certain subpopulations of patients.

 

  5             So, yes, we need to embark on some of

 

  6   these.  We need to realize the limitations as far

 

  7   as predictiveness, and we should not mandate that

 

  8   they become requirements to being the human testing

 

  9   of anti-cancer agents.

 

 10             DR. SANTANA:  Susan?

 

 11             DR. WEINER:  One of the things that

 

 12   occurred to me was that at least some nonclinical

 

 13   data could be very relevant, obviously, to patient

 

 14   selection for trials.

 

 15             DR. SANTANA:  Other comments or issues

 

 16   related to this question?  What I heard, Steve and

 

 17   the rest of the FDA, was that these examples you

 

 18   gave are relevant and, obviously, they are

 

 19   dependent on what you are really after so you can't

 

 20   put them all in one box for each drug.  I think you

 

 21   have to consider them based on each individual

 

 22   agent which is more important in terms of what you

 

 23   want in terms of using preclinical data.

 

 24             You heard my comment about molecularly

 

 25   targeted therapies and potentially how that could

 

                                                               310

 

  1   be an area where some of these models could be

 

  2   used.

 

  3             You heard a little bit also that some

 

  4   agents which potentially may not be totally active

 

  5   but in combination, if you could do that

 

  6   preclinically, you could demonstrate some

 

  7   additional activity before you actually take it to

 

  8   patients.

 

  9             Then I heard comments related to

 

 10   potentially how this could be used to identify

 

 11   potential populations if you could do the

 

 12   preclinical work in animals, looking at some

 

 13   markers that potentially could select the

 

 14   populations that would most benefit once you decide

 

 15   to do the trials.

 

 16             Then the last comment I think came from

 

 17   Peter Adamson that while we do all this, this

 

 18   should not hinder our ability to get the initial

 

 19   clinical pediatric trials started but that they

 

 20   should occur either in parallel or maybe a little

 

 21   bit earlier, or wherever in time, but certainly not

 

 22   to hinder the development even if this data does

 

 23   not exist because, actually, a lot of the questions

 

 24   may come after you do the initial Phase I, some

 

 25   early Phase II studies, and you want to go back to

 

                                                               311

 

  1   certain models and ask the questions that may be

 

  2   relevant by scheduling--are you hitting the right

 

  3   systemic exposure, and things like that.  I think

 

  4   the beauty of this system is that it has to feed

 

  5   back to what you knew from before.  Hopefully, that

 

  6   is something that we will get from this experiment

 

  7   that will be ongoing in the next few years, that

 

  8   information will be used to go back and then ask

 

  9   the relevant questions about why it didn't work so

 

 10   that then, for the next series of experiments, we

 

 11   can potentially address that.  Dr. Helman?

 

 12             DR. HELMAN:  Victor, I want to reiterate I

 

 13   absolutely support what you say, but also just to

 

 14   reiterate what I think both Malcolm and Peter

 

 15   Houghton said which is that, you know, in point of

 

 16   fact this is an experiment.  Many of us have spent

 

 17   our lifetime trying to find better ways to identify

 

 18   screening ways to pick winners for kids and for

 

 19   treating our patients but we don't know.

 

 20             Just as an example and, again, to support

 

 21   what you said and what Peter Adamson said about not

 

 22   mandating or requiring that, I think the GI stromal

 

 23   tumors is a very good case in point.  All we knew

 

 24   is that GI stromal tumors were defined by their

 

 25   mutation in the C KiT receptor.  That was how the

 

                                                               312

 

  1   entity was defined by a group of investigators in

 

  2   Japan, and it allowed us to separate them from what

 

  3   was called up until then GI leiomyosarcomas.  All

 

  4   we knew was that a drug that was active in CML had

 

  5   in vitro activity against C KiT and that was the

 

  6   extent of all the modeling of the data, period,

 

  7   before it was given to a patient with a GI stromal

 

  8   tumor.  The rest is history.  There was no

 

  9   preclinical data.  There were simply two

 

 10   observations, GI stromal tumors had mutations in C

 

 11   KiT and the STI571 AK gleevec had activity in vitro

 

 12   against inhibiting that kinase.  Everything else

 

 13   came later.  So, you know, we were lucky and I will

 

 14   take luck over anything else any day.  So, I think,

 

 15   you know, maybe we will be lucky again.

 

 16             In retrospect, you know, Paul had this

 

 17   data to say that by profiling he could predict, and

 

 18   I would like to hope that in preclinical models we

 

 19   could say that it was absolutely clear that this

 

 20   would have been a winner but we don't know that

 

 21   yet.

 

 22             DR. SANTANA:  Paul?

 

 23             DR. MELTZER:  I just want to make one

 

 24   comment somewhat in the same vein.  I think in

 

 25   pediatric oncology it is extremely important to

 

                                                               313

 

  1   always bear in mind the very large spectrum and

 

  2   number of rare cancers that we encounter and I

 

  3   would not like to see those diseases orphaned from

 

  4   the hope of developing good treatment because we

 

  5   mandate the need for a preclinical model which will

 

  6   never be practical to develop.

 

  7             DR. SANTANA:  Very good point.  I think

 

  8   the practicality of the issues that we have to deal

 

  9   with in tumor systems in pediatric oncology is very

 

 10   relevant to the discussion.

 

 11             DR. DAGHER:  Steven can also address this.

 

 12   I don't think the intent of the question was to

 

 13   imply examples where there would be additional

 

 14   mandates.  I think it was actually in response to

 

 15   issues that have been raised by the cooperative

 

 16   groups themselves and the Phase I Consortium about

 

 17   those kinds of hurdles.  It probably wasn't the

 

 18   intent to ask for additional mandates, although

 

 19   often when FDA asks a question, that is usually

 

 20   what the fear is, that we are thinking about

 

 21   additional mandates.  That wasn't the intent.

 

 22             DR. HIRSCHFELD:  Just to add to Dr.

 

 23   Dagher's precisely right answer, the intent was how

 

 24   can we better inform the data we have?  So, that

 

 25   was the rationale for the entire discussion this

 

                                                               314

 

  1   afternoon, how can we use nonclinical data so that

 

  2   we can improve our conclusions and improve our

 

  3   designs and use our resources most effectively?

 

  4             DR. SANTANA:  Good.  Let's move on to

 

  5   question number two, and I think I am going to ask

 

  6   the FDA to clarify this question a little bit for

 

  7   me, but the question relates to what types of

 

  8   evidence and data would be recommended in each of

 

  9   the following domains to allow extrapolation from

 

 10   nonclinical data and be informative for a clinical

 

 11   condition.  There is pharmacology and

 

 12   pharmacokinetics; safety; efficacy; behavior;

 

 13   long-term effects; developmental aspects; and then,

 

 14   question mark, other domains.

 

 15             Maybe I would like the agency to clarify

 

 16   for me what do they mean by types of data or types

 

 17   of evidence so that we can address this

 

 18   appropriately?

 

 19             DR. HIRSCHFELD:  This is a rather

 

 20   theoretical question but it should be grounded in

 

 21   the limitations of models and should be grounded in

 

 22   data, but there are circumstances where one has

 

 23   information in a domain and would like it to be

 

 24   predictive, or at least informative, for some other

 

 25   domain.  So, in some cases formal rules or formal

 

                                                               315

 

  1   mechanisms have been identified.  As an example,

 

  2   for the conversion from a laboratory measurement

 

  3   from a biomarker to what could be called a

 

  4   surrogate, where the surrogate is for clinical

 

  5   benefit, the NCI and others have made specific

 

  6   recommendations on what type of evidence one would

 

  7   like to see.  Going back about 160 years, there

 

  8   were initially observations which were formulated

 

  9   by Profs. Koch and Henley that there are some

 

 10   conditions that would be met between the

 

 11   identification of a microorganism and its causative

 

 12   role in a disease.

 

 13             So, we don't expect that for all the

 

 14   various domains of clinical interest there are

 

 15   formal rules to be identified, but what we would

 

 16   like to have is some commentary on the type of

 

 17   evidence and the strength of evidence so that if

 

 18   someone is proposing a nonclinical approach we

 

 19   could get some advice on whether we would consider

 

 20   the data that are being offered as valid data, as

 

 21   informative data.  If you want further elaboration

 

 22   we could try, but I think that is the general

 

 23   concept.

 

 24             DR. SANTANA:  If I understood you

 

 25   correctly, I am going to try and see if I follow

 

                                                               316

 

  1   you to contribute to (a).  I think we heard this

 

  2   afternoon how systemic exposures or AUCs of certain

 

  3   drugs can potentially, in certain animal models,

 

  4   predict reduction in tumor volume--not cures but

 

  5   reduction in tumor volumes at the appropriate MTD

 

  6   that are clinically relevant.  So, I think that

 

  7   would be a good example that, if there was good

 

  8   systemic exposure data at the MTD that was

 

  9   clinically relevant in adults that then was going

 

 10   to potentially begin the pediatric studies at that

 

 11   MTD or near that MTD, and there was good response

 

 12   data in animals at that MTD, to me, that would be

 

 13   information that would be relevant to addressing

 

 14   the issue of how pharmacokinetic data could be used

 

 15   in a nonclinical setting in a preclinical kind of

 

 16   model.

 

 17             DR. HIRSCHFELD:  So, if I may paraphrase,

 

 18   and have that then inform the answers to the

 

 19   others, and they may not be the same types of

 

 20   answers, but one has a set of techniques that are

 

 21   available in the nonclinical model that are also

 

 22   available in the clinical model so that one can

 

 23   make direct correlations because the technique for

 

 24   determining AUC is the same in the nonclinical

 

 25   model as in the clinical model and then you are

 

                                                               317

 

  1   relating the readout, applying that technique and

 

  2   then making a direct correlation.  That would be

 

  3   paraphrasing it, but the concept there is that you

 

  4   have techniques which are identical or potentially

 

  5   could map onto each other, and having that assay

 

  6   availability is what lets you make the

 

  7   extrapolation.

 

  8             DR. SANTANA:  Peter and then Donna.

 

  9             DR. ADAMSON:  I think other examples, and

 

 10   it comes back to the need to do tumor biopsies or

 

 11   repetitive tumor biopsies--I think if you can

 

 12   demonstrate in an animal model or, preferentially

 

 13   in animal models, that you have a surrogate that is

 

 14   reasonably predictive of what is happening in the

 

 15   tumor, that should weigh in when looking at the

 

 16   effect in a patient.  So, if you are

 

 17   down-regulating expression of a target in a tumor

 

 18   but you also see it in a lymphocyte and you have a

 

 19   pretty strong correlation in your animal model, it

 

 20   is a lot easier to get lymphocytes from children

 

 21   than it is to get tumors from children.  So, I

 

 22   think that should weigh in as part of proof of

 

 23   principle that you are hitting a target when you

 

 24   actually don't have repeat access to that target.

 

 25             DR. SANTANA:  Donna?

 

                                                               318

 

  1             DR. PRZEPIORKA:  Actually, I would like to

 

  2   ask for additional clarification on this question

 

  3   because I recall one of your first slides in your

 

  4   prior talk was, I believe, that the rule is that

 

  5   you need at least one clinical trial as supportive

 

  6   evidence.  My question is regarding strength of

 

  7   evidence.  Do you want us to be considering

 

  8   sufficient strength of evidence to be the sole

 

  9   supporting data for that one clinical trial because

 

 10   pediatric cancer is an orphan disease and you may

 

 11   not get the chance to do anymore clinical studies?

 

 12             DR. HIRSCHFELD:  Well, if I understood,

 

 13   and we can try to clarify this to be sure we are

 

 14   both addressing the same issue, yes, it is most

 

 15   likely that many pediatric malignancies, for

 

 16   reasons that Dr. Meltzer mentioned, because they

 

 17   are quite rare, will only have one study being

 

 18   done.  Dr. Smith elaborated just on the resources

 

 19   of that too.  So, if we are only going to get one

 

 20   study, there are ways that we can improve our

 

 21   interpretive ability of whatever the clinical

 

 22   outcome may be, either safety or efficacy or

 

 23   long-term effects or something, by using

 

 24   nonclinical data.

 

 25             DR. SANTANA:  Malcolm?

 

                                                               319

 

  1             DR. SMITH:  If there is the one pediatric

 

  2   trial, the one Phase III trial that shows a p value

 

  3   that is favorable and you are looking for something

 

  4   else to help you justify that this is approvable,

 

  5   then looking at a robust preclinical data set that

 

  6   shows the same kind of responses or anti-tumor

 

  7   activity in the preclinical models would seem to be

 

  8   supportive at least and provide you some additional

 

  9   confidence that the agent was going to behave in

 

 10   larger groups of patients as it had in the trial.

 

 11             DR. HIRSCHFELD:  Let me turn it around a

 

 12   little bit.  I guess initially all of you sitting

 

 13   on that side of the room--and since this is an

 

 14   audio recording, it would be Drs. Smith, Helman,

 

 15   Adamson and Anderson--you are starting a fairly

 

 16   extensive program which you acknowledge is an

 

 17   experiment.  So, one way of helping us would be how

 

 18   are you going to know at the end of five years that

 

 19   you have had a successful or an unsuccessful

 

 20   experiment?  And, what are you measuring that is

 

 21   going to determine that?  We would be interested in

 

 22   getting an answer from each of you.

 

 23             DR. SMITH:  I will say something and then

 

 24   let Peter chime in as well.  You know, some of the

 

 25   testing that we do will be to go back and take

 

                                                               320

 

  1   agents that are already being used, for which there

 

  2   is some background response data from the clinical

 

  3   setting, and look at the operating characteristics

 

  4   of the various tumor panels against those agents.

 

  5   So, there will be kind of building of a baseline

 

  6   for agents that we already have activity data for.

 

  7   The others will then be looking ahead

 

  8   prospectively.  If we have agents that have been

 

  9   tested and moved from the preclinical to the

 

 10   clinical setting, is the activity observed

 

 11   preclinically replicated in the clinical setting?

 

 12             DR. ADAMSON:  The clinical endpoints are

 

 13   going to be Phase II endpoints for this experiment,

 

 14   and you have probably heard the reasons why from

 

 15   Malcolm's talk as far as our ability to do Phase

 

 16   IIIs.  But some of those Phase II endpoints are

 

 17   going to be traditional objective response rates or

 

 18   time to progression and I think in part may depend

 

 19   on the agent and our ability to monitor those

 

 20   endpoints.

 

 21             But I should point out also that even in

 

 22   the ideal setting in the next five years where

 

 23   every drug that we potentially want to study will

 

 24   be put through this system, this is not going to be

 

 25   the only path to doing a clinical trial in children

 

                                                               321

 

  1   with cancer.  I can think of a number of

 

  2   circumstances where almost independent of what we

 

  3   see in our model system we are going to be doing

 

  4   clinical studies.  The obvious examples are agents

 

  5   that have remarkable activity in adult cancers.  We

 

  6   are going to look at them in pediatric cancers like

 

  7   we have historically looked in pediatric cancers.

 

  8             And, part of the experiment will be if, in

 

  9   fact, the model predicts lack of activity and we go

 

 10   ahead because of other justifications and find the

 

 11   lack of activity, that is going to also help the

 

 12   negative predictive side of things.  The positive

 

 13   predictive side of things, whether we look at

 

 14   relative response rates of simple yes/no, it met

 

 15   activity thresholds or not, I think will depend

 

 16   upon how many patients and how quickly we can get

 

 17   Phase II trials going.  But there will always be

 

 18   more than one path to get a trial into children

 

 19   with cancer.  The goal, however, will be to put

 

 20   everything that, for whatever reason, has got to a

 

 21   clinical trial through our model system so we can

 

 22   learn both positive and negative predictive values

 

 23   using Phase II as the endpoint.  We would like one

 

 24   day then to start building in toxicity information

 

 25   but right now that is a primary goal of this

 

                                                               322

 

  1   program.

 

  2             DR. SANTANA:  So, if I understood, I think

 

  3   you guys are going to try to address (a) and (c),

 

  4   the pharmacology and pharmacokinetics and efficacy

 

  5   in your models and use that data to decide whether

 

  6   you move on to different model systems or whether

 

  7   you start to introduce other domains, like looking

 

  8   at toxicity and things like that.

 

  9             DR. HIRSCHFELD:  Were there other

 

 10   comments?

 

 11             DR. HELMAN:  Well, again, I maybe would

 

 12   rather address not necessarily the predictive value

 

 13   of the models but the biologic importance of

 

 14   gaining more information.  For example, you know,

 

 15   you heard Malcolm briefly discuss the hope that we

 

 16   can have both some protein profiles, RNA profiles,

 

 17   and if there are subsets--I mean, we are going to

 

 18   use six to ten models so it may be--I have yet to

 

 19   do even a mouse experiment where I consistently

 

 20   cure 100 percent of the mice.  Usually it is 90

 

 21   percent in really good experiments, and sometimes

 

 22   60 percent.  So, if we can identify correlates of

 

 23   response, things that Paul Meltzer talked about,

 

 24   and then find that these are, in fact, important

 

 25   biologic discriminators between people likely to

 

                                                               323

 

  1   respond, for reasons we may have no idea, and just

 

  2   generate hypotheses and if that correlates at all

 

  3   with somehow what we then can use in the clinical

 

  4   study, I think we will make some important steps

 

  5   forward.

 

  6             I would just make the comment that it is

 

  7   something we try to hold ourselves to now because,

 

  8   you know, I think although we all like to think

 

  9   that there are ten more gleevecs out there, the

 

 10   likelihood of hitting a grand slam when we do

 

 11   clinical studies is extraordinarily small.  So, if

 

 12   we do a clinical study with a therapeutic endpoint

 

 13   and the therapeutic endpoint is negative but we

 

 14   learn an important biologic principle, we will

 

 15   continue to make progress.  If the only thing we

 

 16   learn is that this is inactive, we have put a lot

 

 17   of patients into a study that we come out not

 

 18   knowing anything more, other than that this thing

 

 19   is not active.

 

 20             DR. HIRSCHFELD:  Right.  If I may just

 

 21   follow that up, that is exactly the direction where

 

 22   we would like to get some more advice on and

 

 23   thinking.  So, could you elaborate on what you

 

 24   would mean by an important biologic observation

 

 25   even if the clinical result is disappointing?

 

                                                               324

 

  1             DR. HELMAN:  Well, the easiest thing would

 

  2   be we have a kinase that we think is important for

 

  3   the biology of the tumor.  We give a drug.  It

 

  4   inhibits the kinase and all the patients progress.

 

  5   In the end we have learned a very important point

 

  6   which is that that enzyme is irrelevant for the

 

  7   progression of this disease in a patient.  I think

 

  8   that is an incredibly important observation to

 

  9   make.

 

 10             DR. SANTANA:  Dr. Reynolds?

 

 11             DR. REYNOLDS:  I think that one of the

 

 12   things we have to keep in mind when we are talking

 

 13   about these kinds of transitions that you are

 

 14   talking about, Lee and Peter as well, is that the

 

 15   clinical experience in your Phase IIs will be

 

 16   pretty much in patients that are refractory to

 

 17   existing agents.  In some diseases one can imagine

 

 18   that is sort of like up-front patients but for the

 

 19   most part that is patients who have gone through

 

 20   therapy and maybe years out from therapy and it

 

 21   recurred.

 

 22             So, I think in the context of that and

 

 23   thinking about the way the FDA looks at things

 

 24   where they generally approve an agent for a

 

 25   specific indication, like for second-line therapy

 

                                                               325

 

  1   in disease X, we have to keep that in context in

 

  2   the preclinical modeling and we have to make sure

 

  3   that the preclinical modeling doesn't just reflect

 

  4   up-front patients but that it also reflects this

 

  5   refractory population so that we can make those

 

  6   correlations.  For example, what you were talking

 

  7   about, Lee, where you hit your molecular target and

 

  8   you get zero responses, that doesn't mean that the

 

  9   agent wouldn't necessarily work in up-front

 

 10   patients and be an effective agent, and maybe your

 

 11   preclinical models would have said that it worked

 

 12   but then they all developed drug resistance that

 

 13   got around it.

 

 14             So, all those are very complex issues and

 

 15   I think we are going to have to spend a lot of time

 

 16   thinking about these but, more particularly, spend

 

 17   time developing the models so that they reflect the

 

 18   clinical setting as much as we can.

 

 19             DR. SANTANA:  Steve, did you get what you

 

 20   wanted from the panel?

 

 21             DR. HIRSCHFELD:  If I may summarize at

 

 22   least what I heard, and then I will let you, of

 

 23   course, do the more formal summary as we pursue it

 

 24   just a little more because I think this is an

 

 25   important discussion, the context would be that

 

                                                               326

 

  1   people are very interested in nonclinical models.

 

  2   The question is how informative are those data.

 

  3   So, what we have heard so far is that if you have

 

  4   the same technique to measure something, whatever

 

  5   that may be, in the nonclinical model and the

 

  6   clinical model you can do a direct correlation.

 

  7             If you have surrogates in the clinical

 

  8   model that could map onto the nonclinical model,

 

  9   without defining how those surrogates are validated

 

 10   but we will presume that there is a validation

 

 11   process in effect, that could also be used as a

 

 12   mechanism to inform.

 

 13             We also have an approach, to go back to

 

 14   something Dr. Meltzer referred to, training, that

 

 15   we have historical clinical data which then can be

 

 16   used to validate a nonclinical model by using the

 

 17   same types of agents in the nonclinical model and

 

 18   seeing if it correlates to the historical record.

 

 19   So, that is yet another approach.

 

 20             Then we have prospective testing as an

 

 21   approach where we would ask a question of the

 

 22   nonclinical model and ask either the same or what

 

 23   we think is a related question to the clinical

 

 24   model and see if the answer comes out in a way that

 

 25   it is either identical or can be mapped.

 

                                                               327

 

  1             Then, lastly, we have biologic correlates

 

  2   where we are not asking a specific outcome

 

  3   mechanism of the clinical circumstance but we are

 

  4   just trying to pick up information to help

 

  5   mechanistically understand, and then go back to the

 

  6   nonclinical model and use that as some form of

 

  7   evidence.

 

  8             So, that is what we have heard so far, and

 

  9   I think that is all highly useful but, since this

 

 10   is a new area, we want to take the opportunity

 

 11   while we have the expertise available and these

 

 12   presentations fresh in mind to see if there are

 

 13   other aspects that ought to be probed because in

 

 14   some ways we can, hopefully, at least inform if not

 

 15   partially drive a research agenda to improve the

 

 16   validation process.

 

 17             DR. SANTANA:  Dr. Reynolds?

 

 18             DR. REYNOLDS?  Steve, in general what we

 

 19   have been thinking about in terms of when you think

 

 20   about labeling indications and looking for a

 

 21   positive result is to say, okay, this has efficacy

 

 22   in a particular tumor type.  What about the

 

 23   negative condition?  For example, if an agent was

 

 24   to go through clinical trials and show activity and

 

 25   have a registered indication for a pediatric tumor

 

                                                               328

 

  1   but preclinical studies showed that there was a

 

  2   subset of that very disease that was very unlikely

 

  3   to respond to it and there were some limited

 

  4   clinical correlations that showed that was the

 

  5   case, could that be incorporated in the label and

 

  6   used as informative information for pediatric

 

  7   oncologists?  How would the negative side be

 

  8   approached?

 

  9             DR. HIRSCHFELD:  Well, that is exactly one

 

 10   of the scenarios we have been anticipating.  I will

 

 11   give a very brief comment on the aspects of that.

 

 12   First, the question is not restricted just to

 

 13   product labeling.  We are in a position of

 

 14   attempting to advise people on a continuing basis,

 

 15   primarily the pharmaceutical industry but also

 

 16   investigators, saying what type of studies would

 

 17   you like to see?  This is a question that is asked

 

 18   essentially on a daily basis, and all of us spend

 

 19   probably at least 40 percent of our time meeting

 

 20   with people and attempting to answer their

 

 21   questions in this regard.  So, I would view it as

 

 22   the spectrum, and that includes our colleagues

 

 23   whose focus is the domain of nonclinical data.  So,

 

 24   I would view this as a spectrum of how to best

 

 25   utilize resources all throughout the developmental

 

                                                               329

 

  1   cycle of any product and not restrict it just to

 

  2   the labeling.

 

  3             Now, the other aspect is how can we use

 

  4   negative information?  We have used that clinically

 

  5   but I think what you are asking, and this is

 

  6   something that we discussed in April, 2001

 

  7   previously, and that is should negative data inform

 

  8   us to not invest the resources nor expose patients

 

  9   to risk for a given agent?  Now, three years later

 

 10   almost, we would like to ask the question--we are

 

 11   very interested in that because of the potential

 

 12   savings, but what kind of evidence should we use to

 

 13   have confidence in those negative data?

 

 14             DR. REYNOLDS:  If I could just ask Peter,

 

 15   your point being, well, if the agent has some

 

 16   activity some place it should be tested in

 

 17   pediatrics, where could the interface between

 

 18   preclinical model testing that shows it is probably

 

 19   not going to work and limited clinical data in the

 

 20   pediatric setting come together to diminish the

 

 21   number of patients exposed to a potentially

 

 22   ineffective agent?

 

 23             DR. ADAMSON:  As Peter Houghton said, I

 

 24   think until we do this systematically we are not

 

 25   going to be able to answer this question because we

 

                                                               330

 

  1   are just going to have biased data.  So, if we can

 

  2   do it systematically and we can build an experience

 

  3   as far as what these models' positive and negative

 

  4   predictive values are, then I think we really can

 

  5   start making informed decisions when we see

 

  6   negative data that we shouldn't pursue it.

 

  7             Given the limitation of resources, even

 

  8   before we have that data we are likely to apply

 

  9   some of this on an assumption that they are going

 

 10   to be predictive.  But historically, as well as in

 

 11   the current environment, when an agent comes on

 

 12   market for an adult indication it will almost

 

 13   invariably be used by physicians of children who

 

 14   have refractory cancer.  That is the reality.  So,

 

 15   we might as well, for agents that are clearly

 

 16   active and as long as it is not beyond the realm of

 

 17   scientific plausibility--I mean, we are not

 

 18   studying estrogen receptor--well, I shouldn't say

 

 19   that; probably people are--

 

 20             [Laughter]

 

 21             --someone should be able to come up with

 

 22   an example of what wouldn't be used in a child.

 

 23   These drugs are going to be used until we have

 

 24   convincing evidence our models have both positive

 

 25   and negative predictive values.  As Peter said,

 

                                                               331

 

  1   hopefully, in five years we will be able to give

 

  2   you a better answer to that question.

 

  3             DR. HIRSCHFELD:  True enough.  I will just

 

  4   state that we have labeled products that do not

 

  5   have what we consider to be activity in children on

 

  6   the basis of clinical data, sometimes using up to

 

  7   100 children with no evidence of efficacy, at least

 

  8   in a particular disease or particular dose.  We

 

  9   have labeled these things, that they should not be

 

 10   used in children and we are very interested in

 

 11   making sure that there is not inappropriate

 

 12   exposure.

 

 13             DR. SANTANA:  Kind of following that

 

 14   discussion, I think the issue of negative data--you

 

 15   know, it depends on whether you can explain why the

 

 16   data is negative.  That is the critical issue.  It

 

 17   is not that it is negative data because negative

 

 18   data can be very good data.  It is can you explain

 

 19   why it is negative, why it failed?  If you can find

 

 20   the reasons why in your particular experiment it

 

 21   didn't work, to me, that is very informative data

 

 22   and it should not go out with the baby.  You know

 

 23   what I am saying?

 

 24             So, it is a very theoretical discussion of

 

 25   this issue because if you don't do the experiment

 

                                                               332

 

  1   correctly you wind up with negative data, but if

 

  2   you do the experiment correctly and you wind up

 

  3   with negative data and explain why it was negative,

 

  4   to me, that is an advance and I think that should

 

  5   not be thrown out.  Donna?

 

  6             DR. PRZEPIORKA:  Actually, just thinking

 

  7   about Eric's slides from this morning indicating

 

  8   that in the pediatric setting at least we are

 

  9   looking more towards beneficence and doing good for

 

 10   the patient, and having sat on an IRB, I was jut

 

 11   wondering under what circumstances would I get a

 

 12   protocol for a pediatric study that says there is

 

 13   no evidence that this drug is effective in tumors

 

 14   that kids have but we are going to do a Phase II

 

 15   study?  That would be a very difficult protocol to

 

 16   pass through an IRB.

 

 17             DR. ADAMSON:  I agree but there are a lot

 

 18   of protocols that come where there is no data in

 

 19   children.  It is a cytotoxic and there is no data

 

 20   in pediatric models and we do those studies because

 

 21   we accept that cytotoxic agents likely do have

 

 22   activity in pediatric malignancies as a class.  It

 

 23   is a horrendous problem when you think about how

 

 24   little data we base it on.  There has to be some

 

 25   scientific plausibility that the drug is going to

 

                                                               333

 

  1   work.

 

  2             Related to that, I can almost guarantee

 

  3   that gleevec has been tried in every pediatric

 

  4   malignancy to some extent.  What we would much

 

  5   rather do is say let's study it where we think

 

  6   there is scientific plausibility, and we are doing

 

  7   that now on very limited data, basically which

 

  8   tumors do we think express kinases that gleevec

 

  9   might inhibit?  At least that gives us scientific

 

 10   rationale and will give an answer.  If it is

 

 11   negative, I think that is important information

 

 12   because then at least we have the data, we put it

 

 13   out there and people aren't exposing children to

 

 14   gleevec simply because it is the most active agent

 

 15   in CML.  The same is true for adult malignancies as

 

 16   well.  I bet gleevec has been used in virtually

 

 17   every adult cancer that exists by someone.

 

 18             DR. KHANNA:  It is also used in almost

 

 19   every veterinary cancer--

 

 20             [Laughter]

 

 21             -- but one thought I wanted to follow-up

 

 22   with on Peter's comments was that the models are

 

 23   validated or found to be predictive within the

 

 24   context of the agent that was assessed so that

 

 25   agent X with model Y, if there is activity, doesn't

 

                                                               334

 

  1   say that that model is a predictive model for a

 

  2   cancer in general.  So, I think there is a

 

  3   complexity there that has to be incorporated in the

 

  4   next step of the analysis.

 

  5             DR. SANTANA:  Dr. Grillo?

 

  6             DR GRILLO-LOPEZ:  If I may, I would like

 

  7   to focus on the issues at hand in a little bit

 

  8   different way.  Clearly, the medical need that we

 

  9   are discussing is a need to make new effective

 

 10   therapeutic agents available to children as soon as

 

 11   possible.  Now, in the setting of the interaction

 

 12   between the agency and a pharmaceutical company you

 

 13   might look at two extremes.  One extreme might be

 

 14   where an agent is to be developed exclusively for a

 

 15   pediatric malignancy and may not have any

 

 16   applicability in adult malignancy, and those may be

 

 17   very few and far between.  But in that situation I

 

 18   guess the agency has to be more rigorous about the

 

 19   clinical data that needs to be submitted and

 

 20   supported by preclinical data than the other

 

 21   extreme, an agent that is clearly active in adult

 

 22   malignancies and where you could make the

 

 23   extrapolation that it should be active in pediatric

 

 24   malignancies.

 

 25             Most of those agents are the agents that

 

                                                               335

 

  1   we have today in our armamentarium, and most of

 

  2   them have been approved with very little pediatric

 

  3   experience, if any in some cases.  One of the

 

  4   questions is if you do have an agent that is very

 

  5   active and that deserves to be approved for an

 

  6   adult malignancy whose responsibility is it to do

 

  7   the studies to show whether or not it applies in

 

  8   childhood malignancy?  On the one hand, there is

 

  9   the need to find out; on the other hand, there are

 

 10   all of the obstacles that we have discussed today

 

 11   and the fact that there are not enough patients of

 

 12   pediatric age to go around.  We can't do the

 

 13   studies in all of the available agents even today

 

 14   and on the other hand there is the need that we

 

 15   have.  So, as a medical community interested in the

 

 16   cancer patient, we need to find out whose

 

 17   responsibility it is to do those studies.

 

 18             DR. SANTANA:  I think it is all of our

 

 19   responsibility, everybody in this room.

 

 20             DR. GRILLO-LOPEZ:  I think that is the

 

 21   answer.

 

 22             DR. SANTANA:  That is why we are here and

 

 23   we have been here for a long time.

 

 24             DR. GRILLO-LOPEZ:  Let me go further, that

 

 25   answer says that it is not the exclusive

 

                                                               336

 

  1   responsibility of a pharmaceutical company and,

 

  2   therefore, should not be a requirement for approval

 

  3   of an agent that is shown to be active in adult

 

  4   malignancy.  However, how do we approach the issue?

 

  5             The issue can be approached in a variety

 

  6   of ways with the support of the nonclinical data

 

  7   that we have discussed here today, and the simplest

 

  8   way might be to produce clinical and nonclinical

 

  9   evidence that the pharmacology and the

 

 10   pharmacokinetics are similar to those of adults and

 

 11   that the safety profile is similar.  That could go

 

 12   into a package insert without requiring that it be

 

 13   an indication.  Another more stringent way would be

 

 14   to have it go into the package insert of an

 

 15   indication, and there you would require at least

 

 16   Phase II trials as a minimum.

 

 17             DR. HIRSCHFELD:  Rather than addressing

 

 18   the specifics of what goes in product labels and

 

 19   what does not, I would like to summarize by saying

 

 20   it seems that for all of the nonclinical models as

 

 21   they may apply to pediatric oncology we have

 

 22   question marks.  So, I think collectively we should

 

 23   encourage validation and we should encourage

 

 24   multiple approaches to the models so that we can

 

 25   gain confidence in the models and, by gaining

 

                                                               337

 

  1   confidence in the models we can begin to move

 

  2   toward the scenario where the models and the

 

  3   clinical data can be weighted in such a way that we

 

  4   can have a better understanding of what we are

 

  5   looking at.

 

  6             DR. SANTANA:  Yes, I think you said it

 

  7   well.  I think while we move towards perfection, if

 

  8   we could ever reach perfection, the systems that we

 

  9   have at hand have served us to some degree and we

 

 10   should not hinder development of any pediatric

 

 11   studies until those models are truly validation and

 

 12   we have the answers to all the questions.  I think

 

 13   what we have done up to today has served us to some

 

 14   degree and I think the agency needs to recognize

 

 15   that and deal with each one of the drugs or the

 

 16   compounds or the issues at hand on a case-by-case

 

 17   basis, obviously trying to formalize things in such

 

 18   a way so that everybody kind of does it in the same

 

 19   way until we reach that point of perfection.  I

 

 20   think you heard earlier today that it should not

 

 21   hinder our progress until we can validate all these

 

 22   domains and models and come back to you and say

 

 23   this is the best way of doing it.  I don't think we

 

 24   are there yet, and I think that is the difficulty

 

 25   of why we struggle with this question.

 

                                                               338

 

  1             DR. HIRSCHFELD:  Right.

 

  2             DR. SANTANA:  It is very theoretical but

 

  3   we are not there yet.  We can give you some

 

  4   examples but we can't give you the whole universe.

 

  5             DR. HIRSCHFELD:  Clearly.  So, thank you

 

  6   for those examples.  Maybe, in the remaining

 

  7   minutes, we could try to touch on the last

 

  8   question.

 

  9             DR. SANTANA:  Exactly where I was heading.

 

 10   The last question is are there additional

 

 11   recommendations for the effective use of

 

 12   nonclinical data?  For example, will open

 

 13   literature reports be generally acceptable?  Is

 

 14   documentation of compliance with Good Laboratory

 

 15   Practice necessary to evaluate animal data?  Should

 

 16   nonclinical data be submitted as an independent

 

 17   report with a presentation of primary data

 

 18   sufficient for verification and review?

 

 19             I am going to try to skip to the last one

 

 20   and ask the agency how they would use this

 

 21   verification and review when this preclinical data

 

 22   is being presented.  How are you going to judge

 

 23   that data?  It is not just that the data is

 

 24   submitted to you, but what tools and what processes

 

 25   will you use to verify and to review the data? 

 

                                                               339

 

  1   Because I think that will be critical in terms of

 

  2   getting the acceptance of individuals to submit

 

  3   that data--

 

  4             DR. HIRSCHFELD:  Sure.

 

  5             DR. SANTANA:  --whether independent or

 

  6   part of the submission.

 

  7             DR. HIRSCHFELD:  In brief, if we don't

 

  8   have a track record for pediatric oncology it is an

 

  9   open arena so we are attempting to just gain some

 

 10   input into what would be considered acceptable

 

 11   levels of evidence in this regard.  We have much

 

 12   more experience in moving from preclinical to the

 

 13   IND phase, but if we are looking for the

 

 14   nonclinical data to supplement clinical data this

 

 15   would be a new area for us.  So, we don't have

 

 16   precedents and we can't comment to you, for a

 

 17   variety of reasons, about what we would like to

 

 18   see.  We are just trying to get a sense from our

 

 19   invited experts for what you would consider to be

 

 20   acceptable.

 

 21             DR. SANTANA:  Yes, I think the quandary we

 

 22   get into is--

 

 23             DR. GRILLO-LOPEZ:  Clarification, please,

 

 24   acceptable for what?

 

 25             DR. HIRSCHFELD:  Verification of clinical

 

                                                               340

 

  1   findings.

 

  2             DR. GRILLO-LOPEZ:  I am sorry to insist on

 

  3   the clarification.  Although you don't want to talk

 

  4   about labeling but it is an important issue because

 

  5   you could be saying acceptable for labeling.

 

  6             DR. WILLIAMS:  I might elaborate just a

 

  7   little.  I think certainly we do include in our

 

  8   labeling a lot of different pharm tox, biopharm, a

 

  9   lot of different kinds of data and we do accept all

 

 10   kinds of data for clinical use also.  The general

 

 11   principles are that, at least for clinical, we

 

 12   often go out and audit but we have sometimes, in

 

 13   circumstances where we have multiple different

 

 14   literature references that all point to the same

 

 15   thing accept the paper.  Then, as I mentioned

 

 16   earlier, when we get pharm tox data in we generally

 

 17   like to have data to review and generally, if it

 

 18   doesn't meet the GLP standards, we like people to

 

 19   sort of specify how that differs.

 

 20             So, I would sort of maybe even propose

 

 21   that in general those same kinds of standards would

 

 22   probably apply to nonclinical data, that if you

 

 23   didn't do it according exactly to our standards you

 

 24   certainly would support it in some way.

 

 25             DR. SANTANA:  Clarify for me, when a

 

                                                               341

 

  1   sponsor comes to the agency with an NDA and there

 

  2   is preclinical data there, that data gets reviewed

 

  3   and you already have defined what strategies you

 

  4   are going to use to review that data.  What you are

 

  5   implying is that those same parameters would be

 

  6   used for some of these experiments that we are now

 

  7   undertaking.

 

  8             DR. WILLIAMS:  I guess what Steve was

 

  9   saying is when we are talking about a

 

 10   pharmaceutical company that is doing everything

 

 11   under GLP, that is one thing.  It looks like in

 

 12   this setting we might be getting different kinds of

 

 13   data that aren't necessarily exactly as pure as

 

 14   that.  Recognizing that, I guess to what extent

 

 15   would you go to either compromising or specifying

 

 16   in a certain area certain rules or parameters

 

 17   before you would accept it?

 

 18             DR. SANTANA:  Dr. Smith and then Dr.

 

 19   Helman.

 

 20             DR. SMITH:  Certainly for the contract we

 

 21   are involved with, if FDA has recommendations in

 

 22   terms of reports, we would be glad to consider

 

 23   those and to incorporate those and provide you with

 

 24   reports if those are what the agency needed for a

 

 25   particular consideration.

 

                                                               342

 

  1             DR. HIRSCHFELD:  I will just address that

 

  2   before Lee speaks.  We are asking you today for

 

  3   recommendations because we don't have a position

 

  4   yet.  So, that is where we stand.

 

  5             DR. SMITH:  Grant described some kind of

 

  6   characteristics that you might be looking for so we

 

  7   would be open to considering the report formats

 

  8   that would be easier for you to review and be more

 

  9   informative to you.

 

 10             DR. SANTANA:  Lee?

 

 11             DR. HELMAN:  I wanted to ask a question

 

 12   because actually I think it was Dr. Hastings who

 

 13   mentioned this, and nobody has followed up on this

 

 14   and I found it very intriguing, and it follows with

 

 15   some of the information that Chand discussed, which

 

 16   is if we use spontaneous animal models to test the

 

 17   efficacy of a compound and we collected toxicity

 

 18   data, would that be enough if the toxicity data was

 

 19   of high enough quality to not then require

 

 20   additional toxicity data in healthy animals?  In

 

 21   fact, I think there is data to suggest that

 

 22   tumor-bearing animals have toxicity that is not

 

 23   necessarily the same as healthy, normal small

 

 24   mammals.  I mean, it is something we haven't really

 

 25   discussed, which is the coupling of efficacy data

 

                                                               343

 

  1   in pet models and toxicity data, and would that be

 

  2   valid enough to then not require the standard

 

  3   beagle dog or rhesus monkey toxicology?

 

  4             DR. HASTINGS:  Well, first, this is

 

  5   obviously a decision for the oncology division to

 

  6   make about what would be sufficient, but depending

 

  7   on what you knew about the toxicology of a drug to

 

  8   start out with, yes, you might be able to have that

 

  9   as a complete package to support both safety and

 

 10   efficacy.  I think the important issue here

 

 11   though--and this is my own personal opinion and I

 

 12   am not speaking for the division, but what we

 

 13   really would like to have, what I would really like

 

 14   to have is the raw data.  Remember, GLP is

 

 15   basically a set of bookkeeping rules to ensure the

 

 16   integrity of the study and the validity of the

 

 17   data.  That is really what it is all about.  Maybe

 

 18   you won't have a quality assurance statement or

 

 19   anything like that, but I think that is what we

 

 20   would want to have in order to know whether or not

 

 21   the safety data you acquired in a diseased animal

 

 22   model, in fact, is valid enough to make a decision

 

 23   about safety in that condition.  But I think that,

 

 24   yes, you can get toxicity in, as you said, a

 

 25   spontaneous animal model that actually might be

 

                                                               344

 

  1   more relevant to the actual indication than the

 

  2   kind of toxicology data you would get in a healthy

 

  3   animal.  Does that answer your question?

 

  4             DR. HELMAN:  To me, it is really a new

 

  5   concept.

 

  6             DR. HIRSCHFELD:   Our approach is that we

 

  7   will be naive and just for a moment pretend there

 

  8   was no FDA and you don't have to ask us how we want

 

  9   it and you are just trying to make a decision.  So,

 

 10   you have no clinical data and what we are

 

 11   anticipating is that GLP could potentially be a

 

 12   burden on people so you are going to do something

 

 13   less than GLP and you are going to use it yourself

 

 14   to make decisions and to determine whether the

 

 15   model is good or not good.

 

 16             So, what we are asking here is, given that

 

 17   GLP could not necessarily be the standard you could

 

 18   practically adapt, what is the standard that you

 

 19   are comfortable with?  What would you look at; what

 

 20   would you read that you would say, well, this is

 

 21   valid?  So, that is what we are asking.

 

 22             DR. SANTANA:  Dr. Reynolds?

 

 23             DR. REYNOLDS:  Steve, I think the issue,

 

 24   as you hit the nail on the head, is that GLP, which

 

 25   is a very good concept, is not necessarily

 

                                                               345

 

  1   adaptable to the academic setting where limited

 

  2   resources are brought to bear especially on

 

  3   pediatrics where resources are limited.  Whereas

 

  4   the pharmaceutical industry has the investors to

 

  5   spend those resources, we do not necessarily in the

 

  6   academic laboratory have those.

 

  7             The problem is when you say less than

 

  8   that, what would we say is acceptable, well, I

 

  9   think everyone in this room can think of examples

 

 10   from the far end of the spectrum of data where you

 

 11   would ask, well, how did that ever get published

 

 12   all the way to data which according to the

 

 13   regulations is not GLP but is what people would be

 

 14   very confident in using for any purpose.

 

 15             So, what I think we need is not for us as

 

 16   a committee to answer your question, but actually

 

 17   for some guidance from the experts in the FDA that

 

 18   look at GLP issues as to what kind of standards one

 

 19   could apply that are less than full rigor that

 

 20   would be acceptable for the purposes that we want

 

 21   to use these data for.

 

 22             DR. WILLIAMS:  I know that our division

 

 23   commonly accepts things that are not GLP, but we

 

 24   just have the applicant look at the sections of the

 

 25   GLP and tell us how they differ and how they think

 

                                                               346

 

  1   they meet the spirit of it.  So, I think that is

 

  2   doable.  Maybe it could be doable in a more formal

 

  3   setting that met your particular needs for what you

 

  4   are dealing with whether it is tumor models or

 

  5   whatever.

 

  6             DR. REYNOLDS:  Just to finish that, I

 

  7   think this is really important in the concept of

 

  8   what Lee was kind of talking about in terms of

 

  9   using these pet animals, which are fascinating

 

 10   models, because they are never going to make GLP

 

 11   standards.  They are actually clinical practice.

 

 12   So, how does one interdigitate those two different

 

 13   worlds into a process that can then be used by the

 

 14   regulatory process?

 

 15             DR. SANTANA:  Dr. Khanna?

 

 16             DR. KHANNA:  There is a little bit of a

 

 17   precedent that is set for drugs that are pursued

 

 18   for the field of animal health and are approved

 

 19   through the Center for Veterinary Medicine within

 

 20   the FDA.  The issues that we deal with there are

 

 21   basically the availability of raw data, the

 

 22   contemporaneous keeping of records, and the use of

 

 23   standardized tests and measures against those

 

 24   animals.

 

 25             So, speaking only to the use of the pet

 

                                                               347

 

  1   animal studies, there is a body of regulations that

 

  2   oversees these trials and, in fact, those same

 

  3   guidelines which are more GCP-like may be very

 

  4   useful in studies in mice, and they are not as

 

  5   onerous as GLP, and there are probably areas for

 

  6   modification but they may be a good resource to

 

  7   look at.

 

  8             DR. SANTANA:  Dr. Adamson?

 

  9             DR. ADAMSON:  Steve, I think there are

 

 10   really two scenarios.  One is that there are

 

 11   observations made by an independent laboratory that

 

 12   hadn't necessarily set out to generate the data

 

 13   that was going to go to the agency but that is

 

 14   important data.  There, I think the scientific

 

 15   method is a pretty robust one.  That is, it

 

 16   undergoes peer review and if it is important

 

 17   someone ought to repeat it and show the same thing.

 

 18   I would hold any of those observations to the same

 

 19   standards.  I mean, if something is not

 

 20   reproducible by another laboratory, it is not to

 

 21   say throw it away but it should raise some

 

 22   questions.

 

 23             I think what we have been spending more

 

 24   time on is, okay, we are undertaking a program, the

 

 25   sole objective of which is really to provide

 

                                                               348

 

  1   guidance for drug development.  There, I think if

 

  2   you have a standardized approach where the

 

  3   methodology is well described and there are

 

  4   standard operating procedures--again, it isn't GLP

 

  5   but it isn't the opposite of GLP but it is several

 

  6   steps toward it--and I would love to hear Peter

 

  7   Houghton's opinion on this--but I think it would be

 

  8   reasonable to get access to the raw data.  Because

 

  9   usually the limitation of GLP is manpower and

 

 10   resources, and if it is important and we can do a

 

 11   data dump and someone else at the agency can crank

 

 12   through it to see if we get the same results, I

 

 13   think that is a reasonable approach.

 

 14             DR. HIRSCHFELD:  Dr. Hastings, there is a

 

 15   seat there with a microphone and you can take that,

 

 16   and I will just clarify the question.  When we said

 

 17   primary data, that is synonymous with raw data;

 

 18   that is unprocessed data.

 

 19             DR. HASTINGS:  Right.  I just want to make

 

 20   one point.  Actually, we have talked about the

 

 21   pet--well, the companion animal studies.  I believe

 

 22   that under the regulations if you do an

 

 23   experimental study in companion animals or pets you

 

 24   have to have an IND with the Center for Veterinary

 

 25   Medicine.

 

                                                               349

 

  1             DR. KHANNA:  I will just briefly respond.

 

  2   That is not necessarily true.  It depends on the

 

  3   basis around which you are trying to pursue the

 

  4   drug.  If you are pursuing that drug for a

 

  5   veterinary indication it needs to go through the

 

  6   CVM.  If you are not, the CVM has told us that they

 

  7   would not want to be involved in the review of that

 

  8   data that is going towards the human development of

 

  9   a drug.  In fact, they request from us to get

 

 10   regulatory discretion from the human side.

 

 11             DR. HASTINGS:  So, you have already

 

 12   discussed that with CVM?

 

 13             DR. KHANNA:  Yes.

 

 14             DR. HIRSCHFELD:  I can just verify that I

 

 15   was specifically involved in a case or consulted

 

 16   where it turned out to be Dr. Khanna who was

 

 17   submitting a protocol and we were asked whether

 

 18   this was going under an IND that existed for human

 

 19   studies, and we were able to verify that, yes, it

 

 20   was under an IND for human studies and that the

 

 21   data would feed into the collective pool of data

 

 22   for understanding the potential human application

 

 23   and then the Center for Veterinary Medicine

 

 24   gracefully withdrew.

 

 25             DR. WILLIAMS:  It seems like a small

 

                                                               350

 

  1   working group between the Center for Veterinary

 

  2   Medicine and FDA and oncology groups especially

 

  3   could work out some kind of formal/informal

 

  4   arrangement.

 

  5             DR. SANTANA:  Yes, I think that would be

 

  6   critical because as this experiment unfolds over

 

  7   the next few years we want to make sure that the

 

  8   data that we are collecting, and the way we are

 

  9   collecting the data will be acceptable to the

 

 10   agency because, if not, we are going to be faced

 

 11   with the issue of how do we advance drug

 

 12   development in children if the data, for one reason

 

 13   or another, hits a regulatory snarl and is not

 

 14   accepted by the agency.  I think Donna had a

 

 15   question or a comment.

 

 16             DR. PRZEPIORKA:  Yes, for the record, if

 

 17   an academic institution participates in a trial

 

 18   that goes to the FDA, the FDA can come and audit

 

 19   that academic institution to make sure their

 

 20   clinical trial was done appropriately.  If an

 

 21   academic laboratory has their data used to support

 

 22   an IND, is that laboratory open for being audited

 

 23   by the FDA as well?

 

 24             DR. HIRSCHFELD:  The data from that study

 

 25   would be, and we have had circumstances where there

 

                                                               351

 

  1   were, let's say, perceived irregularities in data

 

  2   from a laboratory under a number of INDs and what

 

  3   we have done is for-cause inspections of that

 

  4   facility.  But if it is a single study that an

 

  5   academic laboratory is doing and the data appear to

 

  6   be internally consistent and robust, that usually

 

  7   is not a trigger for an audit.

 

  8             DR. SANTANA:  Dr. Grillo?

 

  9             DR. GRILLO-LOPEZ:  In the setting of an

 

 10   existing NDA, that is a product that has been

 

 11   approved let's say for an adult malignancy, that

 

 12   NDA is a pharmaceutical company's NDA that has been

 

 13   submitted to the FDA and obtained that approval.

 

 14   Usually the way the data works its way into that

 

 15   NDA and the overall database is through the

 

 16   pharmaceutical company.  So, one way that your data

 

 17   would get to the hands of the FDA would be through,

 

 18   in this case, this third party pharmaceutical

 

 19   company that then transmits it to the FDA and the

 

 20   FDA will ask for the raw data.  In fact, in

 

 21   pharmaceutical companies we practically always

 

 22   submit the raw data to the FDA in addition to all

 

 23   analyses and interpretations, etc., with some

 

 24   exceptions where a publication might be sufficient

 

 25   for some particular purpose.  So, in many

 

                                                               352

 

  1   situations you would be working with the

 

  2   pharmaceutical company to put data in a format that

 

  3   would be acceptable to the FDA unless you held your

 

  4   own IND, and then you would file the data to your

 

  5   IND, if I am correct.

 

  6             DR. HIRSCHFELD:  Right, and again we are

 

  7   not restricting it to the NDA filing final phase of

 

  8   development but we are opening the whole discussion

 

  9   to all aspects of product development.

 

 10             DR. GRILLO-LOPEZ:  Yes, I understand but I

 

 11   just wanted to make the point again, as I did

 

 12   earlier this morning, that as we are conducting

 

 13   this discussion it is missing one leg of the stool.

 

 14   I am the only industry representative around this

 

 15   table, and to make this discussion more effective

 

 16   we should have had other industry representatives

 

 17   and presenters who are more expert than I on

 

 18   pediatric oncology.

 

 19             DR. SANTANA:  Dr. Grillo, noted again in

 

 20   the discussion of the afternoon of that issue.

 

 21   Susan, you had a comment?

 

 22             DR. WEINER:  Yes, I guess I wanted to tie

 

 23   it to the discussions earlier in the day from the

 

 24   public's perspective, from the family's

 

 25   perspective.  That is, the most valuable resources

 

                                                               353

 

  1   I think that we all have in this situation are the

 

  2   patient resources, the number of kids who are

 

  3   involved and their well being and time.  Insofar as

 

  4   the conduct of any preclinical or nonclinical

 

  5   activity is done for its own sake or is done

 

  6   without it being in direct service of advancing

 

  7   therapies for kids, I think we have to question

 

  8   that and be mindful of that.

 

  9             In addition, I think it is very important

 

 10   that the agency, when they consider what they

 

 11   require of sponsors or what kinds of studies they

 

 12   believe should be done on kids given what has

 

 13   happened in the preclinical setting, the notion

 

 14   that resources have to be conserved--that risk has

 

 15   to outweigh benefit, to be sure--but that resource

 

 16   have to be conserved because they make commitments

 

 17   into the future that may not be necessary I think

 

 18   is vital, and it is a vital selling point to the

 

 19   families community to hear that from you and I

 

 20   appreciate the exquisite nature with which the

 

 21   discussion is taking place.

 

 22             DR. SANTANA:  I think that is a very good

 

 23   concluding comment for the discussion this

 

 24   afternoon and I couldn't have said it any better.

 

 25   So, unless the agency requests that we provide any

 

                                                               354

 

  1   further comments, I think we have attempted to give

 

  2   you the best that we could, given what we were

 

  3   asked to comment on.  So, I want to thank everybody

 

  4   that participated.  I know it was a tough

 

  5   discussion this afternoon because it was more

 

  6   theoretical based rather than practice based but,

 

  7   hopefully, in the future, once we get more data, we

 

  8   can probably relate it to more practical issues at

 

  9   some future point.

 

 10             DR. HIRSCHFELD:  And I want to thank all

 

 11   of you for helping us.  We could say that we are at

 

 12   the edge and trying to push it but we don't even

 

 13   know where the edge is, and I thank all of you for

 

 14   helping us explore the unknown with the hope that

 

 15   the future will be the known, and our gratitude is

 

 16   noted too.

 

 17             DR. SANTANA:  Thank you, Dr. Hirschfeld.

 

 18             [Whereupon, at 5:10 p.m., the proceedings

 

 19   were adjourned.]

 

 20