1

 

                DEPARTMENT OF HEALTH AND HUMAN SERVICES

 

                      FOOD AND DRUG ADMINISTRATION

 

              CENTER FOR BIOLOGICS EVALUATION AND RESEARCH

 

 

 

            BIOLOGICAL RESPONSE MODIFIERS ADVISORY COMMITTEE

 

                              MEETING #37

 

 

 

 

 

                        Thursday, March 18, 2004

 

                               8:30 a.m.

 

 

                              Hilton Hotel

                        Silver Spring, Maryland

 

                                                                 2

 

                              PARTICIPANTS

 

      Mahendra S. Rao, M.D., Ph.D., Chair

      Gail Dapolito, Executive Secretary

 

      MEMBERS

      Jonathan S. Allan, D.V.M.

      Bruce R. Blazar, M.D.

      David M. Harlan, M.D.

      Katherine A. High, M.D.

      Joanne Kurtzberg, M.D.

      Alison F. Lawton

      James J. Mul, Ph.D.

      Thomas H. Murray, Ph.D.

      Anastasios A. Tsiatis, Ph.D

 

      CONSULTANTS

 

      Jeffrey S. Borer, M.D.

      Susanna Cunningham, Ph.D.

      Jeremy N. Ruskin, M.D.

      Michael E. Schneider, M.D.

      Michael Simons, M.D.

 

      INDUSTRY REPRESENTATIVE

 

      John F. Neylan, M.D.

 

      GUEST HEALTH CANADA REPRESENTATIVE

 

      Norman Viner, M.D.

 

      GUEST SPEAKERS

 

      Stephen Epstein, M.D.

      Silviu Itescu, M.D.

      Robert J. Lederman, M.D.

      Philippe Menasch, M.D.

      Emerson C. Perin, M.D., F.A.C.C.

      Doris A. Taylor, Ph.D.

 

      NIH PARTICIPANTS

 

      Richard O. Cannon, M.D.

      Stephen M. Rose, Ph.D.

 

      FDA PARTICIPANTS

 

      Jesse L. Goodman, M.D., M.P.H.

      Philip Noguchi, M.D.

      Dwaine Rieves, M.D

      Stephen Grant, M.D.

      Richard McFarland, Ph.D., M.D.

      Donald Nick Jensen, D.V.M., M.S.E.E.

 

                                                                 3

 

                            C O N T E N T S

                                                              PAGE

 

      Call to Order

        Mahendra Rao, M.D., Ph.D., Chair                         5

 

      Conflict of Interest Statement

        Gail Dapolito, Executive Secretary                       5

 

      Introduction of Committee                                  9

 

      FDA Opening Remarks

         Presentation of Certificate of Appreciation

         to Retiring Member

         Jesse Goodman, M.D., M.P.H.                            14

         Philip Noguchi, M.D.                                   16

 

                        Open Committee Discussion

                 Cellular Therapies for Cardiac Disease

 

      FDA Introduction and Perspectives

        Dwaine Rieves, M.D.                                     18

 

      Guest Presentations

 

        Overview Cardiomyopathy and Ischemic Heart

           Disease

        Emerson Perin, M.D., Ph.D.                              35

        Q&A                                                     65

 

        Clinical Experience of Autologous Myoblast

        Transplantation

        Philippe Menasch, M.D.                                 85

        Q&A                                                    115

 

        Bone Marrow Cell Therapy for Angiogenesis:

        Present and Future

        Steven Epstein, M.D.                                   128

        Q&A                                                    148

 

      Cellular Therapies for Cardiac Disease

      Richard McFarland, Ph.D., M.D.                           159

 

      Guest Presentations

 

        Myoblasts:  The First Generation Cells for

        Cardiac Repair: What Have We Learned?

        Doris Taylor, Ph.D.                                    169

        Q&A                                                    202

 

        Preclinical Models  - Hematopoietic and

        Mesenchymal Cell Therapies for Cardiac Diseases

        Silviu Itescu, M.D.                                    219

        Q&A                                                    245

 

                                                                 4

 

                      C O N T E N T S (Continued)

 

        From Mouse to Man:  Is it a Logical Step for

        Cardiac Repair?

        Doris Taylor, Ph.D.                                    257

        Q&A                                                    275

 

        Cardiac Catheters for Delivery of Cell Suspension

        Donald Nick Jensen, D.V.M., M.S.E.E.                   292

 

        Transcatheter Myocardial Cell Delivery: Questions

        and Considerations from the Trenches

        Robert Lederman, M.D.                                  307

        Q&A                                                    333

 

      Open Public Hearing                                      343

 

                                                                 5

 

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

 

  2                          Call to Order

 

  3             DR. RAO:  Good morning.  Welcome to the

 

  4   37th meeting of the Biological Response Modifiers

 

  5   Advisory Committee.

 

  6             Today's topic, as you all know, is related

 

  7   to use of cells in cardiovascular disorders, and we

 

  8   have a pretty full schedule for the next couple of

 

  9   days, but before we can start the meeting, we have

 

 10   to have a few sort of committee stuff that needs to

 

 11   be gotten through, so I will turn the mike over to

 

 12   Gail, so that she can make the mandatory

 

 13   announcements.

 

 14                  Conflict of Interest Statement

 

 15             MS. DAPOLITO:  Good morning.

 

 16             The following announcement addresses

 

 17   conflict of interest issues associated with this

 

 18   meeting of the Biological Response Modifiers

 

 19   Advisory Committee on March 18 and 19, 2004.

 

 20             Pursuant to the authority granted under

 

 21   the Committee Charter, the Associate Commissioner

 

 22   for External Relations, FDA, appointed Drs. Jeffrey

 

 23   Borer and Susanna Cunningham as temporary voting

 

 24   members.

 

 25             In addition, the Director of FDA's Center

 

                                                                 6

 

  1   for Biologics Evaluation and Research, appointed

 

  2   Drs. Jeremy Ruskin, Michael Schneider, and Michael

 

  3   Simons as temporary voting members.

 

  4             Based on the agenda, it was determined

 

  5   that there are no specific products considered for

 

  6   approval at this meeting.  The committee

 

  7   participants were screened for their financial

 

  8   interests.  To determine if any conflicts of

 

  9   interest existed, the agency reviewed the agenda

 

 10   and all relevant financial interests reported by

 

 11   the meeting participants.

 

 12             The Food and Drug Administration prepared

 

 13   general matters waivers for participants who

 

 14   required a waiver under 18 U.S.C. 208.  Because

 

 15   general topics impact on many entities, it is not

 

 16   prudent to recite all potential conflicts of

 

 17   interest as they apply to each member.

 

 18             FDA acknowledges that there may be

 

 19   potential conflicts of interest, but because of the

 

 20   general nature of the discussions before the

 

 21   committee, these potential conflicts are mitigated.

 

 22             We note for the record that Dr. John

 

 23   Neylan is participating in this meeting as a

 

 24   non-voting industry representative acting on behalf

 

 25   of regulated industry.  Dr. Neylan's appointment is

 

                                                                 7

 

  1   not subject to 18 U.S.C. 208.  He is employed by

 

  2   Wyeth Research and thus has a financial interest in

 

  3   his employer.

 

  4             With regards to FDA's invited guest

 

  5   speakers and guests, the agency determined that

 

  6   their services are essential.  The following

 

  7   disclosures will assist the public in objectively

 

  8   evaluating presentations and/or comments made by

 

  9   the participants.

 

 10             Dr. Stephen Epstein is the Executive

 

 11   Director, Cardiovascular Research Institute,

 

 12   Washington Hospital Center.  He receives research

 

 13   support, is a consultant to and has financial

 

 14   interests with, firms that could be affected by the

 

 15   committee discussions.

 

 16             Dr. Philippe Menasch is employed at the

 

 17   George Pompidou Hospital in Paris, France.  He has

 

 18   an association with a firm that could be affected

 

 19   by the committee discussions.

 

 20             Dr. Emerson Perin is employed by the Texas

 

 21   Heart Institute.  He receives consultant fees from,

 

 22   and is a scientific advisor to, firms that could be

 

 23   affected by the committee discussions.

 

 24             Dr. Doris Taylor is employed by the

 

 25   University of Minnesota, Center for Cardiovascular

 

                                                                 8

 

  1   Repair.  She receives consultant fees from a firm

 

  2   that could be affected by the committee

 

  3   discussions.

 

  4             Dr. Norman Viner is employed by the

 

  5   Biologics and Radiopharmaceuticals Evaluation

 

  6   Centre, Biologics and Genetic Therapies

 

  7   Directorate, Health Canada, in Ottawa, Canada.

 

  8             FDA participants are aware of the need to

 

  9   exclude themselves from the discussions involving

 

 10   specific products or firms for which they have not

 

 11   been screened for conflicts of interest.  Their

 

 12   exclusion will be noted for the public record.

 

 13             With respect to all other meeting

 

 14   participants, we ask in the interest of fairness

 

 15   that you state your name, affiliation, and address

 

 16   any current or financial involvement with any firm

 

 17   whose product you wish to comment upon.

 

 18             Waivers are available by written request

 

 19   under the Freedom of Information Act.

 

 20             Thank you, Dr. Rao.

 

 21             DR. RAO:  Now you know why I always have

 

 22   Gail read that statement.

 

 23             Before we start any committee work, I

 

 24   would like to welcome two new members to the

 

 25   committee, Dr. Murray and Dr. James Mul.  We

 

                                                                 9

 

  1   generally introduce everyone on the committee

 

  2   first, and we generally go in alphabetical order,

 

  3   but this time I will try and start with the new

 

  4   members, so that they can tell us a little bit

 

  5   about themselves before we have the others

 

  6   introduce themselves.

 

  7                    Introduction of Committee

 

  8             DR. MULE:  I am Dr. Jim Mul.  I am

 

  9   currently the Associate Center Director for the H.

 

 10   Lee Moffitt Cancer Center in Tampa.  I oversee all

 

 11   translational research at the Center including all

 

 12   cell-based therapies for the treatment of cancer as

 

 13   it applies to the clinical treatment of patients

 

 14   with advance tumors.

 

 15             Prior to being in Tampa since September of

 

 16   last year, I was at the University of Michigan

 

 17   Cancer Center for 10 years, and prior to that, the

 

 18   NCI for another 10 years, and I am delighted to be

 

 19   here.

 

 20             DR. MURRAY:  Good morning.  I am Tom

 

 21   Murray.  I am President of the Hastings Center,

 

 22   which is celebrating its 35th years as the world's

 

 23   first research institute devoted to ethics in

 

 24   medicine and the life sciences.

 

 25             I spent 15 years as professor at medical

 

                                                                10

 

  1   schools including 12 at Case Western Reserve

 

  2   University School of Medicine.  My interests are

 

  3   fairly broad.  I write a lot about ethics and

 

  4   ethics in the life science and science policy.

 

  5             Thank you.  I am delighted also to be

 

  6   here.

 

  7             DR. RAO:  If we can go down the table, Dr.

 

  8   Tsiatis.

 

  9             DR. TSIATIS:  Hi.  I am Butch Tsiatis.  I

 

 10   am from the Department of Statistics at North

 

 11   Carolina State University.

 

 12             DR. BORER:  My name is Jeff Borer.  I am a

 

 13   cardiologist.  I work at Weill Medical College of

 

 14   Cornell University in New York City.  I run a

 

 15   division and an institute at Cornell and, relevant

 

 16   to this meeting, I am the Chairman of the

 

 17   Cardiorenal Drugs Advisory Committee of the FDA.

 

 18             DR. CUNNINGHAM:  Good morning.  My name is

 

 19   Susanna Cunningham.  I am a professor in the School

 

 20   of Nursing at the University of Washington in

 

 21   Seattle, and I am the consumer representative for

 

 22   the Cardiovascular Renal Advisory Committee.

 

 23             DR. SCHNEIDER:  I am Michael Schneider.  I

 

 24   co-direct the Center for Cardiovascular Development

 

 25   at Baylor College of Medicine, and our interests

 

                                                                11

 

  1   are in the molecular genetics of cardiac muscle

 

  2   formation, cardiac growth, cardiac cell apoptosis

 

  3   and its relation to heart failure, and, relevant to

 

  4   this meeting, cardiac progenitor cells of different

 

  5   kinds.

 

  6             DR. SIMONS:  Hi.  I am Michael Simons.  I

 

  7   am Chief of Cardiology at Dartmouth Medical School.

 

  8   I work in the area of vascular biology, gene and

 

  9   cell therapy.

 

 10             DR. RUSKIN:  Good morning.  I am Jeremy

 

 11   Ruskin.  I am a cardiologist and

 

 12   electrophysiologist, and I direct the  Cardiac

 

 13   Arrhythmia Service at Massachusetts General

 

 14   Hospital.

 

 15             DR. NEYLAN:  Good morning.  I am John

 

 16   Neylan.  I am a nephrologist and an organ

 

 17   transplanter by training. Currently, I am Vice

 

 18   President of Clinical Research and Development at

 

 19   Wyeth, and I serve as a industry representative to

 

 20   the committee.

 

 21             DR. KURTZBERG:  Hi.  I am Joanne

 

 22   Kurtzberg.  I am a pediatric oncologist.  I direct

 

 23   the Pediatric Bone Marrow and Stem Cell Transplant

 

 24   Program at Duke University and the Carolinas Cord

 

 25   Blood Bank at Duke.

 

                                                                12

 

  1             DR. ALLAN:  Hi.  I am Jon Allan.  I am a

 

  2   virologist at the Southwest Foundation for

 

  3   Biomedical Research.  My area is nonhuman primate

 

  4   models for AIDS pathogenesis.

 

  5             DR. CANNON:  Good morning.  I am Richard

 

  6   Cannon.  I am at the National Heart, Lung, and

 

  7   Blood Institute.  I am Clinical Director of NHLBI,

 

  8   and I am representing NHLBI at this meeting.

 

  9             DR. ROSE:  Good morning.  I am Stephen

 

 10   Rose.  I am Deputy Director for the Recombinant DNA

 

 11   Program in the Office of Biotechnology Activities

 

 12   in the NIH.

 

 13             DR. JENSEN:  Good morning.  My name is

 

 14   Nick Jensen.  I am a reviewer in the Center for

 

 15   Devices and Radiological Health.  I am a

 

 16   veterinarian and an engineer.

 

 17             DR. McFARLAND:  Good morning.  I am

 

 18   Richard McFarland.  I am a reviewer in the

 

 19   Pharm/Tox Branch in the Center for Biologics in the

 

 20   Office of Cellular, Tissue and Gene Therapies.

 

 21             DR. RIEVES:  Good morning.  My name is

 

 22   Dwaine Rieves.  I am a medical officer in FDA's

 

 23   Center for Biologics Evaluation and Research.

 

 24             DR. GOODMAN:  Good morning.  I am Jesse

 

 25   Goodman.  I am the Center Director of the Center

 

                                                                13

 

  1   for Biologics.  I would just like to join in

 

  2   welcoming especially the new members.  My

 

  3   background is as an infectious disease physician in

 

  4   academic medicine for many years.

 

  5             DR. NOGUCHI:  I am Phil Noguchi, Acting

 

  6   Director of the Office of Cellular, Tissue and Gene

 

  7   Therapies in CBER.

 

  8             DR. RAO:  Thank you, everyone.

 

  9             We are very fortunate in having some

 

 10   really leaders in the field come and present some

 

 11   of the data which will be the basis of where we can

 

 12   address some of the questions that have been raised

 

 13   by the FDA.

 

 14             I am going to ask them to just briefly

 

 15   introduce themselves, as well.

 

 16             DR. EPSTEIN:  I am Steve Epstein, a

 

 17   cardiologist.  I am head of the Cardiovascular

 

 18   Research Institute at the Washington Hospital

 

 19   Center.  We are involved in vascular biology, gene,

 

 20   and cell therapy.

 

 21             DR. MENASCHE:  I am Philippe Menasch.  I

 

 22   am cardiac surgeon at the Hospital European George

 

 23   Pompidou in Paris, France.

 

 24             DR. PERIN:  Good morning.  I am Emerson

 

 25   Perin.  I am an interventional cardiologist and

 

                                                                14

 

  1   Director of Interventional Cardiology at Texas

 

  2   Heart Institute in Houston.

 

  3             DR. TAYLOR:  Hi.  I am Doris Taylor.  I am

 

  4   a scientist.  I just moved from Duke University to

 

  5   the University of Minnesota to head the Center for

 

  6   Cardiovascular Repair.

 

  7             DR. ITESCU:  Hi.  I am Silviu Itescu.  I

 

  8   am Director of Transplantation Immunology at

 

  9   Columbia Presbyterian, New York.

 

 10             DR. RAO:  I would also like to welcome Dr.

 

 11   Viner who is from Health Canada.  Health Canada has

 

 12   been following a lot of what the FDA has been doing

 

 13   and it is nice to have them there.

 

 14             I would like to invite Dr. Goodman to make

 

 15   a statement.

 

 16                       FDA Opening Remarks

 

 17           Presentation of Certificate of Appreciation

 

 18                        to Retiring Member

 

 19             DR. GOODMAN:  My main purpose is to thank

 

 20   Joanne Kurtzberg for I guess about four years of

 

 21   service to the BRMAC.  We really appreciate that

 

 22   tremendously.  She has also interacted with CBER

 

 23   before that.

 

 24             One of the reasons I really wanted to come

 

 25   by this morning.  Joanne is rotating off this

 

                                                                15

 

  1   committee.  I know from interactions both within

 

  2   this committee and outside, and from all the

 

  3   leadership and staff within CBER, just what a

 

  4   tremendous advisor and asset Joanne has been for

 

  5   FDA and for your various fields here.

 

  6             Of course, she has mostly contributed very

 

  7   extensively in her areas of hematopoietic stem

 

  8   cells, et cetera, but she has also been a very

 

  9   important thinker and discussant and contributor on

 

 10   the whole range of other cellular therapies and

 

 11   even gene therapy.

 

 12             Please join me in thanking Joanne for her

 

 13   service over these years.  Also, we like to say,

 

 14   particularly CBER, that we are a family and that

 

 15   nobody ever leaves it, and that we, just like a

 

 16   family, we will keep asking for favors in the

 

 17   future and probably causing grief in return.

 

 18             Thanks so much, Joanne.  We have a plaque

 

 19   for her, of course.

 

 20             [Applause.]

 

 21             DR. GOODMAN:  I guess I will just turn it

 

 22   over to Phil to just give a brief introduction for

 

 23   the meeting, but just to say that, as I mentioned a

 

 24   little while back about the islet cell therapies,

 

 25   we, at FDA, are extremely excited about cellular

 

                                                                16

 

  1   therapies and their potential, and I think nowhere

 

  2   is some of that potential clearer, but also perhaps

 

  3   more difficult to evaluate and help move forward

 

  4   than in the area of cardiovascular disease whether

 

  5   it is for ischemic disease or heart muscle disease

 

  6   or trauma, et cetera, some of the uses where there

 

  7   have been some very promising reports.

 

  8             So, we think this is a very timely

 

  9   meeting.  It is very important to get input about

 

 10   how to go forward with efficient development of

 

 11   those products, how to address some of the clinical

 

 12   and safety issues, and how to hopefully make this

 

 13   field positioned to realize its successes in the

 

 14   most efficient manner and also help FDA get that

 

 15   right to the extent that we all can based on

 

 16   incomplete information.

 

 17             Again, we really look forward to this.  I

 

 18   apologize, my usual schedule means I will be in and

 

 19   out,  but I really appreciate it.

 

 20             Phil.

 

 21             DR. NOGUCHI:  Thank you, Jesse, and, of

 

 22   course, Dr. Kurtzberg, our sincere thanks for the

 

 23   many years of service.  Jesse is absolutely right,

 

 24   don't be surprised if the next meeting, you get a

 

 25   funny call early in the morning.

 

                                                                17

 

  1             This is one of our, in a way, continuing

 

  2   series of dealing with things that seem really

 

  3   wonderful and amazing when they come up, where

 

  4   there is a lot of hope and there is perhaps a

 

  5   little bit of hype, but what we have always found

 

  6   over the years, and here I would like to just

 

  7   acknowledge Dr. Rose in the Office of Biotechnology

 

  8   Activities and the Recombinant DNA Committee, what

 

  9   we have learned from them is that one of the best

 

 10   ways that we have of really dealing with things

 

 11   controversial and where there is both hope and

 

 12   there is some trepidation about whether or not this

 

 13   is actually going to work or not, is to bring

 

 14   everyone together, put them in the same room.

 

 15             Our continuing--and this really goes back

 

 16   at least 25 years through the RAC and many years

 

 17   for the BRMAC--is that when you get reasonable

 

 18   people together who may have differing opinions

 

 19   about things, but are presented the facts and the

 

 20   realities, as well as the unknowns, we all

 

 21   basically pretty much come out with the same

 

 22   conclusion, and then we can make significant

 

 23   progress in making these therapies not just

 

 24   experimental, but a reality.

 

 25              With that, what I would really like to

 

                                                                18

 

  1   do, because we have such a full schedule, is now

 

  2   turn it over to Dr. Rieves for the introduction.

 

  3             DR. RAO:  As Dr. Rieves comes up to the

 

  4   mike, I just want to remind people of a few simple

 

  5   rules.  Remember that when you want to ask a

 

  6   question, make sure that you are recognized.  Use

 

  7   the button.  You will see that the light comes on.

 

  8   When you are done, just hit the button again to

 

  9   switch it off, because otherwise, there is sort of

 

 10   a feedback loop and noise.  Make sure you identify

 

 11   yourself when you ask questions.

 

 12              Cellular Therapies for Cardiac Disease

 

 13                FDA Introduction and Perspectives

 

 14             DR. RIEVES:  Good morning.  My name is

 

 15   Dwaine Rieves.  I am a medical officer within FDA's

 

 16   Center for Biologics Evaluation and Research.  This

 

 17   morning I am going to present a brief overview of

 

 18   FDA's perspective on cellular products used in the

 

 19   treatment of cardiac diseases.

 

 20             As will be covered in a subsequent

 

 21   presentation, certain cellular products, when

 

 22   either perfused into the heart or directly injected

 

 23   into heart muscle, are thought to be capable of

 

 24   regenerating heart tissue and/or augmenting heart

 

 25   function.

 

                                                                19

 

  1             Consequently, these products may have

 

  2   special utility in the treatment of heart failure

 

  3   and certain other cardiac diseases.  Today and

 

  4   tomorrow, we will discuss issues in the early

 

  5   clinical development of these products.

 

  6             [Slide.]

 

  7             This talk is divided into three major

 

  8   sections. First, I will cite the purpose in

 

  9   convening this advisory committee.  Secondly, I

 

 10   will provide a regulatory background on FDA's

 

 11   understanding and activities within the realm of

 

 12   clinical development of these products.  Finally, I

 

 13   will introduce the major questions we have proposed

 

 14   for discussion.

 

 15             [Slide.]

 

 16             Unlike many advisory committees where the

 

 17   topics center around assessment of data associated

 

 18   with a specific product or data related to a

 

 19   specific regulatory concern, our purpose in

 

 20   convening this committee is not to obtain

 

 21   definitive regulatory advice, instead, FDA has

 

 22   convened this committee to listen to, and learn

 

 23   from, the voiced thoughts and perspectives with the

 

 24   understanding that this information will enhance

 

 25   our ability to promote the safe clinical

 

                                                                20

 

  1   development of these products.

 

  2             As you are aware, the clinical development

 

  3   of cellular products is in its infancy and many

 

  4   questions surround the very early stages of product

 

  5   development. Consequently, our purpose today and

 

  6   tomorrow is to stimulate a solid scientific

 

  7   discussion of the major facets associated with the

 

  8   very early clinical development of these products.

 

  9             As noted here, we will focus upon three

 

 10   major areas:  manufacturing aspects of the cellular

 

 11   product, preclinical testing of the products, and

 

 12   finally, items related to the early clinical

 

 13   studies.

 

 14             [Slide.]

 

 15             What are the cellular products we will be

 

 16   discussing?  These products may be broadly grouped

 

 17   into two categories.

 

 18             Firstly, those manufactured without

 

 19   ex-vivo culture methodology, that is, the cells are

 

 20   harvested from humans, processed, and then

 

 21   delivered to a recipient without maintaining the

 

 22   cells in culture for a period of time.

 

 23             In general, these cells consist of bone

 

 24   marrow mononuclear cells and certain peripheral

 

 25   blood mononuclear cells, hematopoietic progenitor

 

                                                                21

 

  1   cells that are variously referred to as stem cells,

 

  2   cells thought to be capable of assuming phenotypic

 

  3   characteristics of non-hematopoietic cells.

 

  4             The second category consists of cells

 

  5   that, following harvesting, are maintained in ex

 

  6   vivo culture for a period of time before final

 

  7   processing and administration.

 

  8             In general, these cells consist of those

 

  9   derived from skeletal muscle tissue, cells

 

 10   frequently referred to as myoblasts, and certain

 

 11   bone marrow stromal cells, cells also referred to

 

 12   as mesenchymal cells.  Whether these cultured cells

 

 13   should be regarded as forms of stem cells is more

 

 14   questionable than that for the hematopoietic

 

 15   progenitor cells.

 

 16             Lastly, as the slide notes, most of the

 

 17   cellular products we will be discussing today and

 

 18   tomorrow are of autologous origin.

 

 19             [Slide.]

 

 20             The many questions surrounding the

 

 21   scientific basis for cellular product development

 

 22   illustrate the very nascent nature of the field.

 

 23   As we are probably all aware, there is almost no

 

 24   precedent for the clinical development of products

 

 25   intended to regenerate and/or augment disease

 

                                                                22

 

  1   tissue.

 

  2             The scientific data surrounding this field

 

  3   are relatively new, such that the data are limited

 

  4   in depth and the extent of replication.  Hence we

 

  5   come to the table of clinical development with many

 

  6   hypothetical considerations and some, but

 

  7   relatively limited background supportive data.

 

  8             [Slide.]

 

  9             Given these limitations, our discussions

 

 10   today and tomorrow assume a scientific focus in

 

 11   which certain insights and perspectives are

 

 12   presented, and you, the committee members, will be

 

 13   asked to share your thoughts.  Three points are

 

 14   cited here.

 

 15             First, we acknowledge that these thoughts

 

 16   are all tentative and susceptible to revision based

 

 17   on accumulating data.

 

 18             Secondly, we are not requesting any

 

 19   definitive assessment of data, and we note that the

 

 20   data presented here today are within the public

 

 21   arena, and have not undergone FDA vetting.

 

 22             Finally, I reiterate an earlier comment,

 

 23   that no specific cellular product discussed here is

 

 24   under review with respect to regulatory

 

 25   decisionmaking.

 

                                                                23

 

  1             [Slide.]

 

  2             This slide illustrates the

 

  3   interconnectedness of clinical research and

 

  4   regulatory paradigms.  The connecting link between

 

  5   the two fields is the science.  Clinical research

 

  6   generates the scientific background for clinical

 

  7   development of cellular products and the scientific

 

  8   background forms the major basis for our regulatory

 

  9   paradigms.

 

 10             [Slide.]

 

 11             FDA is charged with many responsibilities,

 

 12   but as cited here, two are especially relevant to

 

 13   this discussion. Specifically, FDA's mission is to

 

 14   promote and protect the public health by optimizing

 

 15   pre-market product development and ensuring

 

 16   sufficient post-marketing product monitoring.

 

 17             The key word in these two statements is

 

 18   "product." A notation that whereas we frequently

 

 19   hear the terms transplant, graft, and procedure, we

 

 20   need to think in terms of a cellular product, a

 

 21   product that is manufactured, labeled, and

 

 22   potentially marketed.

 

 23             [Slide.]

 

 24             A little over 10 years ago, FDA clarified

 

 25   the regulatory basis for oversight of clinical

 

                                                                24

 

  1   development programs for cellular products.  In

 

  2   general, this regulatory framework is the same as

 

  3   that for the drugs and biologic products we

 

  4   commonly recognize as marketed products.

 

  5             Hence, the commonly cited biologic

 

  6   product, drug, and device regulations applied to

 

  7   the clinical development of these cellular

 

  8   products, and the clinical studies must be

 

  9   conducted under the purview of submission of a

 

 10   investigational new drug application.

 

 11             The last bullet on this slide reminds us

 

 12   that clinical development programs may be divided

 

 13   into early and late stages, with the late stages

 

 14   focused upon the ascertainment of data definitive

 

 15   to safety and efficacy, and the early stage, what

 

 16   we are talking about today and tomorrow, focused

 

 17   upon the ascertainment of exploratory safety and

 

 18   bioactivity data.

 

 19             That is, we hope to examine the nature and

 

 20   extent of background data necessary to introduce

 

 21   the cellular products into small, sample size,

 

 22   Phase I clinical studies.

 

 23             [Slide.]

 

 24             As previously noted, the keystone

 

 25   consideration in early clinical development is

 

                                                                25

 

  1   safety.  Specifically, we need to ensure that the

 

  2   tripod of product development is solid. That tripod

 

  3   consists of manufacturing control and testing

 

  4   information, sufficient preclinical testing

 

  5   information, especially information that may inform

 

  6   the design of a clinical study, and finally, the

 

  7   clinical study itself.

 

  8             The next few slides will cite each of

 

  9   these three components.

 

 10             [Slide.]

 

 11             Cellular products must be manufactured in

 

 12   some manner, that is, the cells must be harvested

 

 13   and processed prior to administration to a

 

 14   recipient.  Manufacturing aspects may be divided

 

 15   among four major areas, three being shown on this

 

 16   slide.

 

 17             The top bullet notes that documents should

 

 18   describe the cell source and reagents used in the

 

 19   manufacturing process, such as growth factors,

 

 20   sera, salt solutions and additives.  We need to be

 

 21   confident that all the reagents used in the

 

 22   manufacturing are of clinical or pharmaceutical

 

 23   grade, or that if they are not pharmaceutical

 

 24   grade, they are sufficient for human use.

 

 25             One may envision many potential concerns

 

                                                                26

 

  1   with these materials, such as the use of sera that

 

  2   may contain infections agents, or the use of only

 

  3   partially purified reagents that contain harmful

 

  4   excipients.

 

  5             Secondly, documents should describe the

 

  6   procedures used in manufacturing, specifically

 

  7   describing how cells are aseptically harvested,

 

  8   isolated, and potentially selected.

 

  9             For example, a distinct population of

 

 10   cells may be selected based upon the presence of

 

 11   certain cell surface markers, such as the CD34

 

 12   antigen with the selection process involving

 

 13   incubation with an antibody to CD34.

 

 14             As we know, many investigational

 

 15   antibodies have been developed to target cell

 

 16   surface antigens, and we need to be confident that

 

 17   these selection techniques are performed in a

 

 18   reproducible and safe manner.

 

 19             Additionally, documents should describe

 

 20   the storage and tracking of the cellular products,

 

 21   this being of special concern because certain

 

 22   cellular products may be patient-specific products.

 

 23             For example, measures must be in place to

 

 24   ensure that for autologous products, the cellular

 

 25   product is returned to the correct donor.  Of

 

                                                                27

 

  1   course, the cellular product needs to be labeled as

 

  2   one for investigational use only.

 

  3             The bullet at the bottom of this slide

 

  4   emphasizes the importance of testing the cellular

 

  5   product, an especially important concern since

 

  6   cellular products cannot be sterilized in the same

 

  7   manner as one might sterilize a drug product or a

 

  8   device.  Notable aspects of testing include tests

 

  9   for sterility, endotoxin, viability, enumeration,

 

 10   or cell counting.

 

 11             [Slide.]

 

 12             The fourth component of manufacturing

 

 13   information is product characterization as

 

 14   highlighted here.  When one speaks of product

 

 15   characterization, we are generally talking about

 

 16   cellular phenotype and/or functional

 

 17   characterization and the characteristics of the

 

 18   product's final formulation.

 

 19             For example, a product containing solely

 

 20   CD34 positive cells in saline with no preservatives

 

 21   or media. Product characterization is especially

 

 22   important from a clinical perspective, because

 

 23   failure to consistently manufacture a product makes

 

 24   the clinical data virtually uninterpretable.

 

 25             As noted here, the major aspects of

 

                                                                28

 

  1   product characterization consist of a description

 

  2   of identity, purity, and potency of the final

 

  3   cellular product.

 

  4             [Slide.]

 

  5             Pre-clinical testing is the second major

 

  6   component of product development, and the major

 

  7   aspects of this testing are cited here.  The top

 

  8   bullet notes that consistent with the science, the

 

  9   extent and depth of preclinical testing necessary

 

 10   to support a clinical study is an evolving paradigm

 

 11   and is a major topic for discussion at this

 

 12   meeting.  However, we generally take the stance

 

 13   that this preclinical testing paradigm should be

 

 14   consistent with that used for other biological

 

 15   products.

 

 16             The last bullet notes another important

 

 17   aspect of preclinical testing, the testing of the

 

 18   product administration procedure.

 

 19             This is especially important because many

 

 20   cellular products involve injection directly into

 

 21   heart muscle either through the epicardial surface

 

 22   or the endocardial surface.  These techniques

 

 23   represent inherent safety concerns that may be best

 

 24   evaluated in animals prior to their use in humans.

 

 25             As noted, all available catheters, whether

 

                                                                29

 

  1   marketed or not, are regarded as investigational

 

  2   with respect to administration of cellular

 

  3   products.

 

  4             [Slide.]

 

  5             This slide highlights three aspects of

 

  6   preclinical testing that will be the focus of the

 

  7   preclinical questions tomorrow.

 

  8             Firstly, the choice of the relevant

 

  9   species is central to designing preclinical studies

 

 10   with the major choices being between large animals,

 

 11   such as pigs, versus small animals, such as mice,

 

 12   as well as the choice between immunocompetent

 

 13   animals where, for autologous products, the

 

 14   cellular products would be the animal cells, not

 

 15   human cells, or immunocompromised animals, where

 

 16   the actual human cellular product may be tested.

 

 17             Secondly, designing preclinical studies

 

 18   raise questions of the choice of model, that is, a

 

 19   disease model, such as ischemic heart disease

 

 20   induced in the pig versus a healthy animal.

 

 21             Lastly, preclinical concerns relate to

 

 22   testing of the administration procedure itself,

 

 23   such items as the impact of the catheter materials

 

 24   upon cells, the potential for occlusion of

 

 25   catheters by the cellular product, and the safety

 

                                                                30

 

  1   concerns associated with manipulation of the

 

  2   catheters in the heart.

 

  3             [Slide.]

 

  4             The third component of the clinical

 

  5   development program for cellular products is the

 

  6   clinical study.  There are many aspects of clinical

 

  7   study design that could be discussed, but at this

 

  8   meeting, we are focusing upon two, the first shown

 

  9   here, that is, adverse event detection.

 

 10             This slide highlights two aspects of

 

 11   clinical study design that are frequently

 

 12   engineered to optimize adverse event detection, the

 

 13   evaluation plan with attention to the duration of

 

 14   clinical follow-up, the frequency of evaluations,

 

 15   and the extent or nature of these evaluations.

 

 16             Secondly, the clinical study safety

 

 17   monitoring plan may be optimized through the use of

 

 18   close scrutiny of each study subject based upon the

 

 19   sequential, not simultaneous, enrollment and

 

 20   treatment of the subjects, as well as the

 

 21   prespecifications of the types and numbers of

 

 22   adverse events that should prompt interruption of

 

 23   the study, that is, the study stopping rules.

 

 24             Tomorrow, the committee will be asked to

 

 25   discuss potential adverse events in these early

 

                                                                31

 

  1   clinical studies, both the nature of the events and

 

  2   ways to optimize the safety of the studies.

 

  3             [Slide.]

 

  4             This slide illustrates an additional

 

  5   clinical study design item that we will bring to

 

  6   the committee, that is, a discussion of the

 

  7   analysis of adverse events.

 

  8             Exploratory clinical studies are, by their

 

  9   nature, small sample size studies in which it is

 

 10   often difficult or impossible to distinguish

 

 11   treatment-related events from adverse events that

 

 12   might occur in the natural history of the disease,

 

 13   potential study design mechanisms that might help,

 

 14   but certainly not resolve this issue are cited in

 

 15   the bullets, design features that incorporate

 

 16   randomization of subjects among groups, such that

 

 17   comparisons may be made, the use of controls,

 

 18   especially placebo controls, to make comparisons,

 

 19   the use of masking or blinding to help lessen the

 

 20   bias associated with concomitant therapies or

 

 21   clinical care.

 

 22             Tomorrow, the committee will be asked to

 

 23   discuss mechanisms that might aid in adverse event

 

 24   attribution.

 

 25             [Slide.]

 

                                                                32

 

  1             In this presentation, we have covered

 

  2   three major topics.  Firstly, we have noted that

 

  3   the focus of this meeting is upon a discussion of

 

  4   the scientific aspects of early cellular product

 

  5   development.

 

  6             Secondly, we have noted the regulatory

 

  7   precedent for the cellular products.

 

  8             Finally, we come to the questions.

 

  9             [Slide.]

 

 10             This slide highlights the four major areas

 

 11   of tomorrow's questions.  Specifically, questions

 

 12   related to manufacturing, we will request a

 

 13   discussion of the extent of safety testing and

 

 14   characterization that should be performed prior to

 

 15   the release of a cellular product for

 

 16   administration to humans.

 

 17             The second and third discussion areas are

 

 18   especially critical and may consume the bulk of our

 

 19   time, that is, the extent and nature of preclinical

 

 20   testing necessary to support the introduction of a

 

 21   cellular product into humans, testing that involves

 

 22   questions related to the product itself, as well as

 

 23   the delivery mechanism, the catheter.

 

 24             Finally, we will pose clinical questions

 

 25   centered around adverse event detection and

 

                                                                33

 

  1   analysis with a discussion of the pros and cons

 

  2   associated with the use of controls in these

 

  3   studies.

 

  4             [Slide.]

 

  5             Our agenda is summarized on this slide.

 

  6   As you can see, today, we have a series of invited

 

  7   presentations by FDA staff and leading

 

  8   investigators in the field, as well as the

 

  9   opportunity for public presentations.

 

 10             Tomorrow, we will have another opportunity

 

 11   for public presentations followed by a discussion

 

 12   of the questions.

 

 13             [Slide.]

 

 14             In closing, listed here are some documents

 

 15   that are especially pertinent to our discussions.

 

 16   All these documents are available at www.fda.gov

 

 17   under the CBER sites, specifically the guidance

 

 18   section.

 

 19             The first document is entitled "Draft

 

 20   Guidance for CMC Reviewers: Human Somatic Cell

 

 21   Therapy Investigational New Drug Applications."

 

 22   This document describes the types of information

 

 23   FDA reviewers will examine following the submission

 

 24   of an IND.  Consequently, it provides a very clear

 

 25   description of the types of manufacturing

 

                                                                34

 

  1   information that needs to be submitted with an IND

 

  2   application.

 

  3             The second document is from the

 

  4   International Conference on Harmonization of

 

  5   Regulatory Practices, and it is entitled "

 

  6   Preclinical Safety Evaluation of

 

  7   Biotechnology-derived Pharmaceutics," the S6

 

  8   document.

 

  9             This document is cited because it contains

 

 10   a paradigm that one may apply to cellular products.

 

 11             Finally, the last bullet cites one of the

 

 12   most useful guidances to sponsors and

 

 13   investigators, the ICH Guideline on Good Clinical

 

 14   Practice.

 

 15             This guideline provides detailed

 

 16   information on how to design and conduct a clinical

 

 17   study, information presented in a simple to read,

 

 18   yet relatively comprehensive format.

 

 19             This concludes my presentation and I thank

 

 20   you for your attention.

 

 21             [Applause.]

 

 22             DR. RAO:  Before we continue with the rest

 

 23   of the presentations, I would like to just welcome

 

 24   Dr. Harlan and ask him to introduce himself.

 

 25             DR. HARLAN:  I apologize for being late,

 

                                                                35

 

  1   but I am David Harlan, NIDDK.  I study

 

  2   transplantation of islets and immunotherapies.

 

  3             DR. RAO:  Our first speaker will be Dr.

 

  4   Perin, whom you already were introduced to.

 

  5                        Guest Presentations

 

  6        Overview Cardiomyopathy and Ischemic Heart Disease

 

  7             DR. PERIN:  I want to thank you for the

 

  8   invitation to be here to present to you today,

 

  9   especially Dr. Grant, who has helped me put this

 

 10   together in a way.

 

 11             So, what I want to do here this morning,

 

 12   the task that has been laid before me is that of in

 

 13   a way setting the stage or giving you a general

 

 14   idea of the kinds of patients that we are treating.

 

 15             Obviously, this is fundamental if we are

 

 16   thinking about doing clinical trials.  It is very

 

 17   important to understand the nature of the disease

 

 18   in which these kind of therapies will frequently be

 

 19   applied.

 

 20             What I plan to do is talk about the

 

 21   following topics.  First, we will start from the

 

 22   beginning, define what heart failure is, look at

 

 23   the scope of heart failure, talk a little bit about

 

 24   the pathophysiology, look at some prognostic

 

 25   markers, talk about the treatment to some extent 

 

                                                                36

 

  1   and that is important in terms of monitoring, and

 

  2   then really work our way towards end stage heart

 

  3   failure because that is where I think the focus of

 

  4   most of the future clinical trials will likely be

 

  5   initially, and finally, talk about adverse events,

 

  6   which I think is a major concern, and the

 

  7   monitoring of there adverse events.

 

  8             Now, I know many of you are not

 

  9   cardiologists, so hopefully, I can go from a level

 

 10   where we are not getting too complicated, but not

 

 11   too simple.

 

 12             Starting with the definition of what heart

 

 13   failure is.  Firstly, heart failure is a clinical

 

 14   syndrome very simply defined by certain symptoms

 

 15   and certain signs that come together.  These

 

 16   symptoms are fatigue, shortness of breath, and

 

 17   congestion, and these are translated on a physical

 

 18   exam by being able to hear a third heart sound, the

 

 19   patient manifesting peripheral edema, and jugular

 

 20   venous distention.

 

 21              If we start looking at this problem and

 

 22   have a broad overview of this, first, I want to

 

 23   show you a graph from the HOPE trial.  This is a

 

 24   trial that was conducted in thousands of patients,

 

 25   as you can see here, over 9,000 patients.  It was a

 

                                                                37

 

  1   study primarily of ramipril and vitamin E in

 

  2   patients with hypertension over a long period of

 

  3   time, involved a five-year follow-up.

 

  4             But it is just very interesting, as we

 

  5   start out looking at heart failure, to look at this

 

  6   patient population, and here we have over 500 days,

 

  7   so here is about a year out, and if we look at this

 

  8   population, who is not primarily designated as

 

  9   particularly sick or harboring heart failure, that

 

 10   identified the patients that did have heart failure

 

 11   and we look at their survival, you will see the

 

 12   mortality.

 

 13             It separates from the beginning, and when

 

 14   we get out to about a year, you have got a 10

 

 15   percent mortality in the group that has heart

 

 16   failure compared to less than 4 percent mortality

 

 17   in the general population.  So, you can see that

 

 18   the problem that we are dealing with seems to be

 

 19   very serious.

 

 20             If we go here and let's just look at the

 

 21   placebo arms of some very large heart failure

 

 22   trials, these are trials pretty much aimed at

 

 23   evaluating different forms of therapy now in heart

 

 24   failure patients, and looking at different severity

 

 25   of heart failure patients, for example, in the

 

                                                                38

 

  1   V-HeFT trial, inclusion criteria might be an

 

  2   ejection fraction less than 40 percent.

 

  3             If we look at PRAISE, which evaluated

 

  4   amlodipine in more severe heart failure, an

 

  5   ejection fraction was less than 30 percent,

 

  6   comparing this with Class III and Class IV

 

  7   patients, very sick patients.

 

  8             So, you can see here if we look at just

 

  9   the placebo arms of all these trials, a very

 

 10   striking mortality as we go along.  If we look at 1

 

 11   year here, this will vary from 10 percent down to

 

 12   around 30 percent.

 

 13             If we go out to 2 years in the very sick

 

 14   patients, we see that half of the patients are

 

 15   dead.  So, heart failure, depending on the

 

 16   presentation, carries a very ominous prognosis.

 

 17             It is a very broad problem, 5 million

 

 18   Americans are living with heart failure now,

 

 19   550,000 new cases are diagnosed each year.

 

 20             From 1979 to 2000, heart failure deaths

 

 21   increased by 148 percent.  Now, what is

 

 22   interesting, over this period of time, we have

 

 23   actually gotten a lot better at treating heart

 

 24   failure, and we do treat it. I will get into this a

 

 25   little later, and I will show you the modern treat

 

                                                                39

 

  1   of heart failure and how much better we are doing,

 

  2   but at the same time that we are treating heart

 

  3   failure better, we are also treating the patients

 

  4   that have coronary disease, which is a very

 

  5   dominant problem in this country and around the

 

  6   world,  we are treating those patients better, too,

 

  7   so what happens is we are getting more patients

 

  8   with heart disease that normally would have died

 

  9   earlier, to live longer, and as we are able to

 

 10   bypass and stent and do all these revascularization

 

 11   procedures and come up with better treatments, we

 

 12   are getting people that go further down the road,

 

 13   that otherwise would have succumbed a long time

 

 14   ago.

 

 15             So, despite our improvements in treatment

 

 16   of coronary disease, we are dealing with an

 

 17   increasing amount of heart failure deaths.

 

 18             In individuals diagnosed with heart

 

 19   failure, cardiac death occurs at 6 to 9 times the

 

 20   rate in the general population.  If you are more

 

 21   than 40 years old, you have a 1 in 5 chance of

 

 22   developing heart failure, and 22 percent of men and

 

 23   46 percent of women that have heart attacks will be

 

 24   disabled within 6 years with heart failure.

 

 25             So, as you can imagine, the high

 

                                                                40

 

  1   prevalence and multiple complications have an

 

  2   implication in terms of health costs.  If we look

 

  3   at the costs, and these numbers vary, and it

 

  4   depends on what you are looking at and what year

 

  5   you are looking at, but this is a very significant

 

  6   financial burden on the country, over 5 percent of

 

  7   the total health care costs.

 

  8             You can see that most of the cost involved

 

  9   is really involved in inpatient care, and as I will

 

 10   show you hopefully, that really translates to the

 

 11   sickest portions of these patients, that as you get

 

 12   sicker with heart failure, you start coming into

 

 13   the hospital more, and that is what runs up the

 

 14   cost of treating these patients.  It is interesting

 

 15   that transplant is just a little sliver out of the

 

 16   pie here.

 

 17             So, let's look at the causes of heart

 

 18   failure, and I am not going to get into all the

 

 19   little minor details, but let's look at the major

 

 20   causes of what brings on heart failure.

 

 21             Seventy-five percent of people that go on

 

 22   to develop heart failure had hypertension

 

 23   previously.  Valvular heart disease is a big

 

 24   contributor and also heart failure engenders

 

 25   valvular heart disease, mitral regurgitation

 

                                                                41

 

  1   further contributes to the problem.

 

  2             Coronary artery disease, you are all

 

  3   familiar with this, the number one problem in this

 

  4   country, and this is really what we are going to

 

  5   focus majorly on in terms of causing heart failure

 

  6   and the specific kind of heart failure that this

 

  7   engenders.

 

  8             In cardiomyopathy, there is many different

 

  9   kinds of things that get a heart to perform poorly,

 

 10   all the way from an idiopathic cardiomyopathy to

 

 11   such things as iron overload, et cetera, which are

 

 12   not as common.

 

 13             Now, what I want to talk about here is

 

 14   really systolic heart failure.  There is something

 

 15   called diastolic heart failure, and that really has

 

 16   a lot to do with compliance problems of the

 

 17   ventricle, and in these patients, we are going to

 

 18   see a normal ejection fraction.

 

 19             So, this is really a different animal and

 

 20   it is really not what we are focusing on, so what I

 

 21   am going to be talking about today is systolic

 

 22   heart failure, and as I will show you, with the

 

 23   hallmark being a low left ventricular ejection

 

 24   fraction.

 

 25             This is just to give you a practical

 

                                                                42

 

  1   example.  This is an angiogram from one of the

 

  2   patients that we treated with stem cell therapy in

 

  3   Brazil, who all had an ejection fraction that

 

  4   averaged about 20 percent.  This patient has an

 

  5   ejection fraction of 10 percent.

 

  6             You can see the coronaries are calcified.

 

  7   This is a catheter in the left ventricle.  This

 

  8   heart is supposed to be pumping this contrast we

 

  9   just put into the aorta.  As you can see, it is not

 

 10   doing that very well at all.  Only 10 percent of

 

 11   what is in here gets out with each beat.

 

 12             So, you can tell this is a dilated big

 

 13   heart that just doesn't contract well.  That is the

 

 14   picture of severe heart failure right there, and

 

 15   this is what I want to talk about.

 

 16             Now, when we talk about heart failure, I

 

 17   think everybody is aware of the classification.

 

 18   There is Class I, II, III, IV, which are commonly

 

 19   used, but it is important to acknowledge this.

 

 20   Class I involves no limitation of physical

 

 21   activity, Class II slight limitations, Class III

 

 22   marked limitations, you can't walk up a flight of

 

 23   stairs without getting short of breath, and Class

 

 24   IV, you have symptoms at rest.

 

 25             If we look at this, if we put Class III

 

                                                                43

 

  1   and Class IV together, you see the division is

 

  2   about a third for each of these pieces of the pie

 

  3   here.

 

  4             Now, if somebody comes in with Class IV

 

  5   heart failure, they are very short of breath at

 

  6   rest, you can give them some diuretics and they

 

  7   will feel better.  They are not Class IV anymore,

 

  8   they are Class III.

 

  9             So, it is interesting, there has been a

 

 10   want in development of a little different way of

 

 11   looking at heart failure, and a staging or

 

 12   classification put out by joint AHA and ACC shows

 

 13   four different stages, and really looks at heart

 

 14   failure more like a disease like cancer.

 

 15             So, where we can identify patients that

 

 16   are at high risk of developing it, we can screen

 

 17   patients, and then we can start treating patients

 

 18   before they really manifest symptoms of the

 

 19   disease.

 

 20             Again, this is a progressive disease and

 

 21   we are going to end up with people that are

 

 22   refractory even to all kinds of treatment.  I am

 

 23   going to go over this a little bit more in detail a

 

 24   little later.

 

 25             So, in defining what heart failure is, I

 

                                                                44

 

  1   hope I have given you a general idea of the scope

 

  2   of the problem, just talk a little bit about what

 

  3   causes it because it is important to understand

 

  4   that to be able to know how we treat it and how we

 

  5   monitor these patients.

 

  6             Usually, we are talking about ischemic

 

  7   heart disease and we are dealing with a myocardial

 

  8   insult, which is usually a heart attack, so that

 

  9   heart attack causes damage to the heart muscle, and

 

 10   that is going to result in dysfunction of that

 

 11   heart muscle.

 

 12             Well, the body is going to try to

 

 13   compensate this dysfunction and especially in two

 

 14   major ways.  One is neurohumoral activation, so we

 

 15   will talk a little bit about this in more detail,

 

 16   but essentially, these compensatory mechanisms are

 

 17   going to make the heart change its shape and its

 

 18   size.  It is something we call remodeling.  It

 

 19   involves hypertrophy of the myocytes and then it

 

 20   involves fibrosis and dilatation.

 

 21             So, these mechanisms that the body helps,

 

 22   to try to help to reverse what is going on,

 

 23   actually wind up causing toxicity, hemodynamic

 

 24   alterations that all lead to remodeling, and

 

 25   remodeling really is the hallmark.

 

                                                                45

 

  1             You saw that big heart.  Well, remodeling

 

  2   is how you get from a normal small heart, which you

 

  3   have, to a big boggy heart that doesn't contract.

 

  4   That is the problem of heart failure.

 

  5             This was very simply put by Doug Mann in a

 

  6   nice editorial a few years ago.  Basically, here is

 

  7   the heart over time, as we have an index event, and

 

  8   basically, remodeling occurs, the heart gets

 

  9   bigger, the ejection fraction goes down as time

 

 10   goes by and symptoms occur as time progresses, as

 

 11   well.

 

 12             So, I have told you we have a myocardial

 

 13   insult. This leads to LV dysfunction and

 

 14   remodeling, and this really instigates a

 

 15   neurohumoral response.  In return, this is going to

 

 16   have an impact on remodeling again.

 

 17             So, what are these neurohumoral things

 

 18   that happen?  Well, first of all, most importantly,

 

 19   is the renin- angiotensin-aldosterone system, and

 

 20   there are several points in which the body

 

 21   upregulates the system and ultimately, it acts on

 

 22   the AT-1 receptor, which will cause

 

 23   vasoconstriction, proteinuria, again LV remodeling.

 

 24             As you can identify, here are several

 

 25   sites in which medications, the mainstay of some of

 

                                                                46

 

  1   the therapy for heart failure works, namely ACE

 

  2   inhibitors that work at this point, ARBs that work

 

  3   at this point, beta blockers have a role in

 

  4   inhibiting renin, as well.  So, some of the

 

  5   mainstay of therapy is actually directed at one of

 

  6   these mechanisms of compensation.

 

  7             On the other side, we have sympathetic

 

  8   activation. We have increased sympathetic activity

 

  9   that again leads to myocardial toxicity and

 

 10   arrhythmias, and then on the other side, with the

 

 11   sympathetic outflow, we get vasoconstriction. This

 

 12   impacts negatively on the kidney, sodium retention,

 

 13   more vasoconstriction, and progression of the

 

 14   disease.

 

 15             Just to get a slightly little bit more

 

 16   complicated, just to mention that it is really not

 

 17   all that simple, there are other things involved,

 

 18   and we have cytokines, TNF-alpha, IL-6,

 

 19   inflammation that actually progresses with the

 

 20   progression of heart failure.

 

 21             Endothelin is a potent vasoconstrictor.

 

 22   All these things lead to apoptosis and unfavorable

 

 23   effects upon the myocyte, but then lead to LV

 

 24   remodeling, which I have told you is one of the

 

 25   mainstays of reasons for heart failure.

 

                                                                47

 

  1              Now, natruretic peptides are important,

 

  2   as well. It's another compensatory mechanism that

 

  3   the body has.  I am sure you are familiar with

 

  4   these BNP, it's a B-type natruretic protein that

 

  5   actually comes from the ventricle, the A types

 

  6   comes from the atrium.  We will just focus on the B

 

  7   type.

 

  8             What this does, basically, in response to

 

  9   elevated pressure inside the heart, we secrete BNP.

 

 10   This suppresses the renin-angiotensin-aldosterone

 

 11   system and suppresses endothelin.  It helps with

 

 12   peripheral vascular resistances, decreases

 

 13   vasodilatation, and it increases natruresis.

 

 14             So, if we go on to understand now that

 

 15   there is an interplay between LV dysfunction and

 

 16   remodeling, and that basically, this will lead to

 

 17   low ejection fraction, and that is what we see in

 

 18   the patients.

 

 19             On the other hand, as a result of this, we

 

 20   will start getting a constellation of symptoms, and

 

 21   it is the combination of having a low ejection

 

 22   fraction and symptoms that defines heart failure.

 

 23             Let's look a little bit at the prognostic

 

 24   markers. I just talked a little bit about BNP.

 

 25   Well, it is very interesting.  If we divide BNP in

 

                                                                48

 

  1   quartiles here, depending on the amount of BNP that

 

  2   you have circulating, your survival will go down.

 

  3   It is a prognostic marker, as well as a treatment.

 

  4   Norepinephrine, the same way.  So, these are

 

  5   markers of prognosis.

 

  6             It is very interesting.  These are levels

 

  7   of BNP, and if you can decrease them, decrease to a

 

  8   less degree, or here, we have an increase.  So,

 

  9   depending on which direction your BNP goes, your

 

 10   survival varies as well, and that is an important

 

 11   concept.

 

 12             Let's look at another different kind of

 

 13   marker. Exercise capacity, peak oxygen consumption.

 

 14   In the transplant world, this is very important.

 

 15   Here you see the number 14, so a peak oxygen

 

 16   consumption greater than 14 or less than 14 has

 

 17   very different prognostic indicators and in many

 

 18   centers, this serves as a marker threshold for one

 

 19   of the criteria for entering the patient into a

 

 20   transplant program.

 

 21             You can see here a difference in mortality

 

 22   from 53 percent mortality over two years in

 

 23   patients that have an NVO2 of less than 14, to that

 

 24   of 11 with greater than 14, so this is another

 

 25   important number in patients with heart failure.

 

                                                                49

 

  1             Then, if we look overall and look at

 

  2   symptoms and hospitalizations, here is a New York

 

  3   Heart Class I to IV, and this is fairly intuitive,

 

  4   but as we get more symptomatic, we have an impact

 

  5   on survival, and as we are getting more

 

  6   symptomatic, we have an increase in

 

  7   rehospitalization.

 

  8             What about ejection fraction?  I just

 

  9   talked about ejection fraction, and you can see

 

 10   here, similarly to NVO2, ejection fraction can

 

 11   divide prognostically how patients will do.  Here

 

 12   we see more than 20 percent, less than 20 percent.

 

 13   Here you see a two-year survival, 54 percent, so

 

 14   half the people dying that have an ejection

 

 15   fraction less than 20 percent.  At one year, that

 

 16   is a little over 20 percent.

 

 17             The same thing, this is a large randomized

 

 18   clinical trial, ejection fraction less than 40

 

 19   percent.  Over time, people die more frequently.

 

 20             Now, let's add a little arrhythmia to

 

 21   this.  Looking at different levels, the first two

 

 22   are greater than 30 percent ejection fraction, here

 

 23   less than 30 percent, so that stratifies that out,

 

 24   but then if we just add the amount of extra

 

 25   ventricular beats to this, and if we have less than

 

                                                                50

 

  1   10 per hour, more than 10 per hour, and then with a

 

  2   poorly contractile ventricle, your survival goes

 

  3   down as we add extra ventricular beats.

 

  4             One attempt that has been made to sort of

 

  5   graph this problem, because now I have shown you

 

  6   many different prognostic markers and different

 

  7   things we can use to classify these patients to

 

  8   decide what to do and how to follow them.

 

  9             One of them is a heart failure survival

 

 10   score.  There is an invasive model, there is a

 

 11   non-invasive model.  So, things like cause of heart

 

 12   failure, resting heart rate, EF, mean blood

 

 13   pressure, if there is a conduction delay

 

 14   electrically in the heart, oxygen consumption, and

 

 15   serum sodium can enter into a risk classification.

 

 16             Here, you just basically have a graph that

 

 17   shows according to low, medium, and high, your

 

 18   survival will vary according to the risk.

 

 19             In our little schema here, that leads

 

 20   symptoms and low ejection fraction to heart

 

 21   failure, what are really the things, though, that

 

 22   are driving mortality?  They are going to be pump

 

 23   failure, on the one hand, and arrhythmia, on the

 

 24   other, because sudden death, as I talked to you

 

 25   about before, is a very prominent problem in people

 

                                                                51

 

  1   that have heart failure.

 

  2             So, it is the combination of these three

 

  3   things that will pretty much drive patients to a

 

  4   lethal exit.

 

  5             Let's talk a little bit about treatment

 

  6   now.  What are the goals of treatment of heart

 

  7   failure?  You want to delay the progression or

 

  8   reverse remodeling, which you can do in some

 

  9   patients, and delay the progression and reverse

 

 10   myocardial dysfunction.

 

 11             You want to reduce mortality, relieve the

 

 12   symptoms, improve functional capacity, and reduce

 

 13   disability, also decrease the intensity of medical

 

 14   care and hopefully reduce economic cost.

 

 15             I have shown you we go from initial

 

 16   injury, initial infarct, we suffer remodeling, we

 

 17   get a remodeled heart that now has a low ejection

 

 18   fraction, and over this course of time, we have a

 

 19   worsening of symptoms, so how are we going to

 

 20   impact this in terms of treatment?

 

 21             Well, the two mainstays are neurohumoral

 

 22   blockade, we have kind of gone over some of the

 

 23   things that we can do, and we will look at those,

 

 24   and the other is revascularization.  So, many times

 

 25   with the use of medication or with the use of

 

                                                                52

 

  1   revascularization, we can reverse some of this

 

  2   remodeling in some patients, and in some patients

 

  3   we don't.

 

  4             One thing that is very important in terms

 

  5   of being able to recover patients that have

 

  6   remodeled hearts, and that are in this road of

 

  7   heart failure, is identification of viable

 

  8   myocardium.

 

  9             Myocardial viability has clearly been

 

 10   shown to influence the prognosis of people that are

 

 11   undergoing revascularization procedures, so if you

 

 12   have a viable myocardium, you are going to do

 

 13   better.  You have a chance of improving more than

 

 14   someone who doesn't.

 

 15             Just to shift gears for just a second

 

 16   here, these are electromechanical maps.  These are

 

 17   representations of the left ventricle.  This is

 

 18   from a patient in our Brazil stem cell study.

 

 19             This is an electrical map, this is a

 

 20   mechanical map.  Let's just look at the electrical

 

 21   map because I just talked to you about viability.

 

 22   Very simply, if your cells are alive, they have an

 

 23   electrical signal that is high.  If you have a big

 

 24   scar with no cells, you have no electricity, you

 

 25   have a low electrical signal.

 

                                                                53

 

  1             We put it on a little color scale.  Red is

 

  2   dead or red is very little voltage.  Purple is

 

  3   high.  Here, you see on this electromechanical map,

 

  4   an area of myocardial viability.  Again, just as it

 

  5   is important to understand viability when you are

 

  6   vascularizing patients that have heart failure,

 

  7   that have coronary disease, it is also going to be

 

  8   important, in my view, to understand myocardial

 

  9   viability when we are applying some of these

 

 10   therapies, and I think there will be differences in

 

 11   bone marrow therapies and myoblast therapy, but

 

 12   that is something to keep in mind.

 

 13             I just wanted to show you an example of

 

 14   the very common things that we deal with, so this

 

 15   is not some esoteric difficult patient to find.  We

 

 16   come across people like this all the time in the

 

 17   hospital every day.

 

 18             This is a patient who was 41 years old, he

 

 19   had bypass, he stopped up all his vein grafts and

 

 20   his memory artery, and he had ejection fraction of

 

 21   20 percent, very similar to the one that I showed

 

 22   you, and Class IV congestive heart failure.

 

 23             This gentleman was really delightful.  He

 

 24   was actually a pilot for a major airline, and

 

 25   because of his bypass, he had to be put off the

 

                                                                54

 

  1   flying, and he was actually in charge of all the

 

  2   simulators, and he was the guy that graded all the

 

  3   pilots when they had to come in and do the

 

  4   simulation testing.

 

  5             Basically, here, we have a 41-year-old

 

  6   guy, very active man who has gone bypass, he has

 

  7   lost his graft, he obviously has very aggressive

 

  8   disease, and why I hear the talk about why some

 

  9   people have more aggressive coronary disease than

 

 10   others.

 

 11             You see this is his right coronary, it is

 

 12   completely blocked up, X's mean that you can't see

 

 13   anything on angiography, so this kind of fills from

 

 14   the other side by collaterals, see these little

 

 15   twigs down here.

 

 16             Then, the circumflex is completely

 

 17   occluded.  This is a floating marginal branch.

 

 18   This is supposed to be connected, but this is

 

 19   totally occluded, as well.  The only artery he has

 

 20   got left is the one down the front of his heart,

 

 21   but this is very much infarcted, and has a very

 

 22   significant blockage here, as well as the takeoff

 

 23   of this.

 

 24             So, this patient, there is really nothing

 

 25   to do, and we are faced with this a lot every day. 

 

                                                                55

 

  1   This patient, as I have shown you these curves of

 

  2   mortality, this patient at our hospital wound up

 

  3   going for an LVAD type procedure and died, and that

 

  4   is what we see again and again, so this is a very

 

  5   serious problem.

 

  6             So, looking of an overview of treatment of

 

  7   heart failure, let's see, we have medical-based

 

  8   therapy, on one hand, we have device-based therapy,

 

  9   on the other.

 

 10             On the medical side, we need neurohumoral

 

 11   blockade, we can have a hemodynamic approach and

 

 12   also antiarrhythmic approach, so we are going to

 

 13   use these drugs, ACE inhibitors, aldosterones,

 

 14   diuretics, beta blockers, and then antiarrhythmics,

 

 15   such as amiodarone, and then we are going to use

 

 16   more potent i.v. inotropes that improve

 

 17   hemodynamics, and asaratide [ph], which is

 

 18   basically similar to BNP, it is like giving the

 

 19   patient BNP.

 

 20             On the other hand, we are going to have a

 

 21   device-based approach using resynchronization

 

 22   therapy.  It really hasn't shown a benefit in

 

 23   survival, but in combined endpoints.  We are going

 

 24   to put defibrillators into people, and I will show

 

 25   you how that has improved survival.

 

                                                                56

 

  1             Then, we will have ventricular assist

 

  2   devices, and when all this fails, we have an option

 

  3   of heart transplant, that is very little available

 

  4   actually, and as you saw, it is a very little

 

  5   sliver of what we are able to do.

 

  6             But as you cumulatively add these

 

  7   therapies, you are able to impact on survival and

 

  8   make patients live longer.  Here, you see sort of

 

  9   adding digoxin and diuretic, adding an ACE

 

 10   inhibitor, and then adding a beta blocker, we get

 

 11   progressive improvement.  So, this is pretty well

 

 12   established in terms of medical therapy.

 

 13             When we look at defibrillators, here is a

 

 14   curve. This is from the MADA-2.  This is primary

 

 15   prevention, defibrillator in patients, previous MI,

 

 16   LVF less than 30 percent, a very significant

 

 17   survival difference in the patients that get a

 

 18   defibrillator, so treating the arrhythmias is also

 

 19   important.

 

 20             Back to our schema of the different

 

 21   classification of stages of heart failure.  You see

 

 22   that we can gradually, we start with ACE inhibitors

 

 23   and gradually add different medications, but

 

 24   everybody kind of goes up these stairs and ends up

 

 25   here at the top, and that is why we have increasing

 

                                                                57

 

  1   mortality from heart failure, because we are

 

  2   getting people to get to this point where before

 

  3   they really didn't reach that stage.

 

  4             Then, we get to a stage of basically

 

  5   refractory symptoms, so they have been bypassed,

 

  6   they have had stents, everything has been done for

 

  7   them, and they have that bad heart, it doesn't pump

 

  8   well, they have a lot of symptoms, they can't

 

  9   breathe very well.  Many of them have angina.  I

 

 10   want to want to give you a little bit of my own

 

 11   perspective on that.

 

 12             If we look at current trends, this was

 

 13   published last week in JACC, very interesting.

 

 14   Heart failure treatment--this is the survival

 

 15   curves--heart failure treatment in 1994 to 1997.

 

 16   Here is a survival curve.  We have improved the

 

 17   treatment of heart failure.

 

 18             1999 to 2001, gee, we are doing a lot

 

 19   better, and this is comparable actually to

 

 20   transplant from 1993 to 2000, and it really raises

 

 21   the question if transplant, with the modern

 

 22   management in medical management of heart failure,

 

 23   how important is it and what the role of transplant

 

 24   really is.

 

 25             Really, there is a gap between a very

 

                                                                58

 

  1   invasive transplant or LVAD and the medical

 

  2   therapy, there really is, and we are here to talk

 

  3   about stem cell therapy.  There is a gap of

 

  4   something that could be done that is not quite as

 

  5   invasive and traumatic as an LVAD or transplant,

 

  6   and that can improve the patient significantly

 

  7   since we are doing so well with medical therapy.

 

  8             I want to talk to you a little bit about

 

  9   my perspective on end-stage ischemic heart disease.

 

 10   Basically, as I have told you, we have improved the

 

 11   medical management, so we have longer survival, we

 

 12   have improved the vascularization treatments of

 

 13   coronary disease, we have improved the survival

 

 14   following a heart attack, and that is why we have

 

 15   more patients, and now we are using widely

 

 16   defibrillators, and that is why people are living

 

 17   longer.

 

 18             So, this is sort of my understanding of

 

 19   this end-stage patient.  You progress with coronary

 

 20   disease until you get to the Stage III and Stage

 

 21   IV, Class III/Class IV heart failure.

 

 22             If we look at these patients, sometimes

 

 23   there will be a little surprise, because some

 

 24   patients really just have shortness of breath, so

 

 25   this is a variable.  This may occupy the whole

 

                                                                59

 

  1   square or angina may occupy the whole square.

 

  2             So, some patients predominantly have heart

 

  3   failure, and these patients that predominantly have

 

  4   heart failure probably weren't very good at forming

 

  5   collaterals when they had heart attacks and

 

  6   developed a lot of scar tissue, and have a very low

 

  7   ejection fraction.  These are the sickest patients

 

  8   and the patients that are going to have a very high

 

  9   mortality.

 

 10             On the other hand, but also in the Class

 

 11   III or Class IV, and sometimes we pool these people

 

 12   together in trials and that is why I am making this

 

 13   distinction, some people have angina more than they

 

 14   have heart failure.  These probably have a much

 

 15   better collateral formation when they had these

 

 16   events, so their ejection fraction is a little more

 

 17   preserved.

 

 18             I have had many patients that have lived

 

 19   on one artery.  Their whole heart is beating okay.

 

 20   That one artery feeds everything by collaterals,

 

 21   but they are in really bad shape.  I mean it's an

 

 22   illusion that they are doing okay, but they do have

 

 23   a preserved ejection fraction, and their

 

 24   manifestation is a lot of chest pain.

 

 25             So, symptoms can vary from one side to the

 

                                                                60

 

  1   other and some patients have a balance here, and I

 

  2   think we need to keep this in mind when we are

 

  3   designing these trials.

 

  4             So, there is a predominant angina, and

 

  5   this is the kind of patient that got, let's say,

 

  6   these TMR type procedures.  That is the kind of

 

  7   population you are dealing with.  The predominant

 

  8   aspect is disabling angina, preserved EF, 100- to

 

  9   200,000 new cases per year, and constitute about 5

 

 10   percent of the patients undergoing angiography at

 

 11   tertiary referral centers.  This has been studied

 

 12   in this particular case at the Cleveland Clinic.

 

 13             One year mortality is still very high.

 

 14   Then, that other group, predominantly heart failure

 

 15   symptoms, very low EF, myocardial ischemia, though,

 

 16   is still present, but with more scar.  No option

 

 17   really for any kind of revascularization.  One year

 

 18   mortality, 20 to 50 percent.  I have shown you one

 

 19   curve where it is up to 80 percent, I mean it can

 

 20   be really bad.

 

 21             Here, we have ICD therapy trials.  If we

 

 22   look at secondary prevention trials, very sick

 

 23   patients in this study, treated with amiodarone,

 

 24   you see here one year mortality 44 percent.  I mean

 

 25   heart failure can be worse than cancer.

 

                                                                61

 

  1             Here is the REMATCH trial.  This is an

 

  2   LVAD.  This is the impact of LVAD, and there is an

 

  3   impact of survival, but again you are dealing, in

 

  4   this case, with Class IV patients that are

 

  5   unresponsive to medical therapy, so these very sick

 

  6   patients, but again an invasive, costly, not widely

 

  7   available kind of therapy, but it does have an

 

  8   impact on failure.

 

  9             I want to finish now talking a little bit

 

 10   then, hopefully, I have given you an overview of

 

 11   the problems with heart failure, and how are we

 

 12   going to look at adverse events.

 

 13             Well, what are the things that are going

 

 14   to drive the adverse events here, are going to be

 

 15   arrhythmia, ejection fraction, and symptoms, and I

 

 16   think if we focus here, we can pretty much decide

 

 17   what we need to look at in these patients over time

 

 18   as we use new therapy towards these patients.

 

 19             Let's look at low ejection fraction, how

 

 20   are we going to monitor that?  Well, we need to

 

 21   look at cardiac function, cardiac size, and the

 

 22   perfusion status of the ventricle.  We can do that

 

 23   very simply, if you take a simplistic approach,

 

 24   with echocardiography.

 

 25              I empirically have placed this here based

 

                                                                62

 

  1   on my own limited experience here, but I read in

 

  2   the document that you wanted some more practical

 

  3   advice, so I will give you my own sort of practical

 

  4   feel for what I would do.

 

  5             If we did echocardiogram on these

 

  6   patients, we could do it monthly for the first

 

  7   three months and then at six months follow-up.  We

 

  8   can do SPECT, we know that we don't need it too

 

  9   early, and that is a very simple way of doing it,

 

 10   three to six months.  Clinical visits, which will

 

 11   be very frequent, and I will talk about that, and

 

 12   BNP can be done for that, as well.

 

 13             Now, we can get fancy and use alternative

 

 14   imaging strategies, we can use MRI,

 

 15   electromechanical mapping, PET, depending on the

 

 16   institution, and depending on what we are really

 

 17   looking for and want to find.

 

 18             Cardiac arrhythmias, it is important to

 

 19   monitor cardiac rhythm.  Holter monitoring is very

 

 20   simple, probably should be done after the

 

 21   procedure, one, three, six months later.  Q-T

 

 22   interval when the patient comes in for his clinic

 

 23   visit is a strong predictor of survival, just a

 

 24   plain-old, good-old 12-lead EKG, and that should

 

 25   always be looked at.

 

                                                                63

 

  1             In the patients I guess that are getting

 

  2   myoblast therapy, there may be a little bit more

 

  3   concern about this, and this is really not my area

 

  4   of expertise, but these patients, many of them

 

  5   already entering with an AICD, that have sort of a

 

  6   built-in little computer that is already monitoring

 

  7   their rhythm as it is.  If they don't, you might

 

  8   want to consider event monitoring.

 

  9             For symptoms, well, clinical visits

 

 10   biweekly for 8 weeks, monthly up to 6 months.  We

 

 11   are going to look at heart class, we are going to

 

 12   look at EKG, CBC, CRP, look for inflammation.

 

 13   Exercise capacity, ramp treadmills, as you know, if

 

 14   you put a patient that has end-stage heart failure

 

 15   on a graded treadmill test, every time the

 

 16   treadmill bumps up and goes a little faster, he

 

 17   just may not be able to exercise at that point.

 

 18             So, the advantage of a ramp treadmill

 

 19   protocol is that you have a gradual continuous

 

 20   increase, so these people that really can't do very

 

 21   much at all, they will be able to tolerate the

 

 22   exercise and probably get further than they could

 

 23   in any other kind of exercise test.

 

 24             There is a very simple way of evaluating

 

 25   an exercise test, a 6-minute walk test.  You just

 

                                                                64

 

  1   define a distance, walk the patient walk for 6

 

  2   minutes, see how fast he can go.  You can do that

 

  3   at a clinic visit, and it is very simple to do.

 

  4   So, you can do something like this at one, three,

 

  5   and six months.

 

  6             Rehospitalization.  We look at the

 

  7   rehospitalization rates.  It is important to look

 

  8   at the use of i.v. medications that are used to

 

  9   control symptoms, because this is, as you saw, the

 

 10   biggest part of the pie in terms of costs, and is a

 

 11   real problem in the end-stage patients.

 

 12             Quality of life, it is important to assess

 

 13   quality of life, for example, SF36, Minnesota

 

 14   Questionnaire.

 

 15             Just some suggestions.  I want to wrap

 

 16   this up and saying I hope I have given you a

 

 17   general idea and scope of this problem.  We deal

 

 18   with a very, very serious problem, which is heart

 

 19   failure, specifically, that which is ischemic heart

 

 20   failure and specifically, end-stage ischemic heart

 

 21   failure.

 

 22             I hope I have given you a flavor of this

 

 23   and set the stage for the discussions.

 

 24             Thank you very much.

 

 25             [Applause.]

 

                                                                65

 

  1             DR. RAO:  Thank you, Dr. Perin.

 

  2             There is time for questions, and we can

 

  3   open it up to the committee.

 

  4                               Q&A

 

  5             DR. SCHNEIDER:  Emerson, one of the things

 

  6   that you did very nicely was lay out the clinical

 

  7   spectrum for people who may not be familiar with it

 

  8   in this context.

 

  9             I wanted to follow up on that point

 

 10   because work presented at international meetings

 

 11   recently by the Frankfurt group of Andreas Sire and

 

 12   Stephanie Dimler suggests that bone marrow derived

 

 13   cells and circulating progenitor cells from

 

 14   patients with established heart failure may be

 

 15   deficient relative to the performance of bone

 

 16   marrow derived and circulating progenitor cells

 

 17   from patients with an acute infarct.

 

 18             So, while it is not quite an apples and

 

 19   oranges comparison to envision cardiac cell

 

 20   grafting immediately post infarction or in the

 

 21   first week post infarction in patients without

 

 22   severe ventricular dysfunction versus patients,

 

 23   let's say, two to four months out with mild or no

 

 24   ventricular dysfunction versus the end-stage heart

 

 25   failure patients who have been a focus in your talk

 

                                                                66

 

  1   this morning, it does seem to me that that clinical

 

  2   heterogeneity introduces a couple of problems.

 

  3             I am curious to know how you have worked

 

  4   those through in your own work.  One of them is

 

  5   because what we are discussing today and tomorrow,

 

  6   is autologous cell therapy, I believe that there is

 

  7   a serious issue of patient-to-patient cell

 

  8   heterogeneity which has been relatively little

 

  9   discussed in the field except in these still

 

 10   unpublished or perhaps one paper has come out in a

 

 11   secondary journal from Stephanie and Andreas about

 

 12   the defects.

 

 13             So, one question is what kinds of

 

 14   standards should a proposed production center be

 

 15   required to meet in terms of their ability to

 

 16   generate cells that perform in accordance with some

 

 17   standard when there is patient-to-patient variation

 

 18   of this kind.

 

 19             Secondly, if you are envisioning putting

 

 20   cells of different kinds into a so severely an

 

 21   ischemic background as the 41-year-old former pilot

 

 22   that you mentioned, doesn't it become important to

 

 23   clearly distinguish, as the prefatory remarks did,

 

 24   between mechanisms of action for proposed donor

 

 25   cells that are aimed at regeneration specifically

 

                                                                67

 

  1   versus benefits that are achieved through entire

 

  2   different mechanisms, such as angiogenesis?

 

  3             If you put new cells into an ischemic

 

  4   background, they will surely die, and if the goal

 

  5   is to achieve angiogenesis in a background where

 

  6   the native coronary circulation has failed and the

 

  7   graft has failed, then, it seems to me we need a

 

  8   clearer resolution of the problem of which cells do

 

  9   which things well, and really fine-tune much better

 

 10   than the field has to date, you know, which are the

 

 11   cells that we want where the spectrum is normal

 

 12   vasculature, insufficient muscle cells versus the

 

 13   hypothetical ischemic patient that you described

 

 14   where revascularization is the major goal.

 

 15             DR. PERIN:  Well, that's fantastic.

 

 16             [Laughter.]

 

 17             DR. PERIN:  I think the basic answer to

 

 18   your question is I don't know, but, you know, these

 

 19   are all very good points, starting with the cell

 

 20   type, we really don't know.

 

 21             Actually, we have submitted a manuscript

 

 22   in which we have had the pathology of one or our

 

 23   patients in our study in Brazil who received

 

 24   autologous bone marrow, died 11 months later, and I

 

 25   really can't preempt I guess our publication, but I

 

                                                                68

 

  1   think we will be seeing some evidence of myogenesis

 

  2   and angiogenesis from autologous bone marrow cells,

 

  3   but we really don't know what we are getting when

 

  4   we are putting, let's say, autologous bone marrow,

 

  5   and even in that patient that has, let's say he has

 

  6   predominantly ischemia, if we want to

 

  7   revascularize, can we get a predominantly

 

  8   angiogenic effect, so we really don't know, and we

 

  9   need to define that.

 

 10             Mononuclear fraction of the bone marrow is

 

 11   a very simple approach, the one that we have taken,

 

 12   and it seems to initially, and we haven't really

 

 13   done efficacy studies and we are continuing on, but

 

 14   there is a suggestion that it does, so I think that

 

 15   we need to take every step that we take should be

 

 16   put one foot in front of the other, and if  the

 

 17   mononuclear cell fraction works, I think we can go

 

 18   from there and keep investigating that.

 

 19             Now, the average age in our trial was

 

 20   about 58, and you mentioned the problem--

 

 21             DR. RAO:  Can I interrupt?  These are

 

 22   really important questions, but they discuss data

 

 23   which was not presented in the talk right now.  I

 

 24   would like to at least focus the questions

 

 25   initially on the issues that relate to the

 

                                                                69

 

  1   presentation right now.

 

  2             We should really come back to these

 

  3   questions tomorrow when we discuss exactly these

 

  4   sorts of issues.

 

  5             Do you think that that would be okay with

 

  6   you, Dr. Schneider?

 

  7             DR. SCHNEIDER:  We will certainly return

 

  8   to them tomorrow, but I was discussing issues that

 

  9   were raised in this talk, which was clinical

 

 10   heterogeneity.

 

 11             DR. RAO:  Let's then focus, not on the

 

 12   cells per se, and the choice of cells, because none

 

 13   of the presentation was related to the production

 

 14   facility or how the cells would be, or the quality

 

 15   would be, or how you would choose the mechanism,

 

 16   but maybe how do you choose patients for a trial or

 

 17   is there some reasonable way of selecting patients,

 

 18   that there would be consensus on.

 

 19             DR. PERIN:  Okay.  So, we will get back to

 

 20   your first question and really, that is something

 

 21   that actually, we are working on trying to

 

 22   understand, is there a thumbprint or is there a

 

 23   profile in the study by Dimler and their colleagues

 

 24   looking at the characteristics of cells in certain

 

 25   patients, and obviously, they may not be the same

 

                                                                70

 

  1   in a diabetic, in a severe heart failure, we don't

 

  2   know, so there is another important we don't know.

 

  3             Age obviously is a very important thing,

 

  4   so harvesting cells from a 75-year-old may be very

 

  5   different than doing that in a 55-year-old, so

 

  6   these are all questions that need to be answered.

 

  7             DR. RAO:  Dr. Mul.

 

  8             DR. MULE:  Given the slides you showed of

 

  9   the steps toward progression of heart failure, and

 

 10   given the current interventions along that pathway,

 

 11   from your perspective, where would you see

 

 12   cell-based therapy intervention falling into that

 

 13   step toward complete heart failure?

 

 14             DR. PERIN:  Right now, at close to the

 

 15   last few steps, I think ethically, we are propelled

 

 16   to really study the problem in the patients that

 

 17   really don't have a proven conventional option for

 

 18   treatment.  In brief, I would say in the patients

 

 19   who can't be revascularized, because really medical

 

 20   therapy, we are going to apply to everyone, so then

 

 21   we are left with revascularization.

 

 22             Well, can we revascularize?  Well, we do,

 

 23   and we do it again and again, and there is a point

 

 24   where you are out of revascularization options, and

 

 25   I think that is one  place we are initially now,

 

                                                                71

 

  1   then, you could think about applying this kind of

 

  2   treatment.

 

  3             DR. HARLAN:  Building upon what Dr. Rieves

 

  4   mentioned when he gave his introductory comments, I

 

  5   want to just congratulate you on, it seems like our

 

  6   task is to weigh the risk-benefit, and you have

 

  7   outlined very clearly the risk, and I accept that

 

  8   it is severe, and I also want to congratulate you

 

  9   on mentioning the JACC paper that was just

 

 10   published, that showed how dangerous it is to look

 

 11   at historical controls, because we are making such

 

 12   rapid progress.

 

 13             My question is along those lines, not in

 

 14   this field, I just read in the journal, the

 

 15   Washington Post, about the great advance that has

 

 16   been made in super-high statin therapies, and I

 

 17   wonder if you could comment on that study, that

 

 18   these super-physiologic statin doses seem to have a

 

 19   major impact on mortality.

 

 20             DR. PERIN:  I really don't have an

 

 21   expertise in a lot of things, and that is not one

 

 22   of them, so it is really hard for me to comment on

 

 23   that.  I know that it looks like giving people HDL

 

 24   in the future may be a very exciting thing, and we

 

 25   may be able to finally find our liquid plumber kind

 

                                                                72

 

  1   of solution for people.

 

  2             Then, again, statins are just--more and

 

  3   more if you study statins, you have probably come

 

  4   to the conclusion it should be in the water pretty

 

  5   soon, I mean the patient benefit is on every single

 

  6   aspect of cardiovascular disease.

 

  7             DR. RAO:  Dr. Kurtzberg.

 

  8             DR. KURTZBERG:  You mentioned some

 

  9   practice-based methods to evaluate outcomes and

 

 10   function in these patients, but I think the

 

 11   challenge is to determine what the cells are doing,

 

 12   you know, are they differentiating into other kinds

 

 13   of cells, are they mediating inflammation, are they

 

 14   mediating angiogenesis, and I don't see how you can

 

 15   sort that out by clinical-based study.

 

 16             Do you know of other technologies that are

 

 17   on the  horizon that may help with that, that are

 

 18   non-invasive, or would you consider serial biopsies

 

 19   in patients like this to answer those questions?

 

 20             DR. PERIN:  That is a good question.  I

 

 21   don't know that serial biopsies would be a very

 

 22   efficient way of evaluating that.  You would have

 

 23   to have a very precise way of being able to

 

 24   identify where you put the cells and be able to go

 

 25   exactly to that same spot.

 

                                                                73

 

  1             We do have that technology.  Dr. Lederman

 

  2   is going to follow me eventually here.  The MRI

 

  3   field, I think is very promising in that regard in

 

  4   terms of labeling and following cells.

 

  5             Now, I really don't know that even

 

  6   labeling a cell, even if it died, if the label

 

  7   stays there,  you still see the label, so I think

 

  8   that we have to even go a step further and be able

 

  9   to prove the functionality of the cell that is

 

 10   alive and was implanted.

 

 11             That can be done on an experimental basis,

 

 12   so we figure ways out to do that, but this is a

 

 13   very intriguing problem and a very difficult

 

 14   problem to evaluate.  I think  you have put your

 

 15   finger on something that is going to be hard to

 

 16   know.

 

 17             DR. DINSMORE:  Jonathan Dinsmore from

 

 18   GenVec.

 

 19             I just had a question on your angina heart

 

 20   failure continuum.  I was confused because most

 

 21   heart failure patients present without angina, with

 

 22   symptoms of fatigue, so what percentage of heart

 

 23   failure patients actually experience angina?

 

 24             DR. PERIN:  If we are talking about

 

 25   ischemic heart failure, we are not talking about

 

                                                                74

 

  1   other kinds of heart failure, actually, idiopathic

 

  2   heart failure, you kind of get the same remodeling

 

  3   and everything except you didn't have that infarct

 

  4   in the beginning, but you go through the same sort

 

  5   of pathophysiologic processes.

 

  6             So, we are talking about ischemic heart

 

  7   failure. People that have ischemic heart failure

 

  8   have coronary disease.  Coronary disease is

 

  9   narrowing of your coronary arteries.

 

 10             Depending on what your response is, you

 

 11   will or will not have angina, but angina is one of

 

 12   the manifestations of coronary disease, and it is

 

 13   really not a good thing to base a lot on, because

 

 14   the expression of angina is very variable.

 

 15             It depends on your pain threshold.  I mean

 

 16   if you are a diabetic, you may not have as much

 

 17   pain.  It is a subjective thing subject to

 

 18   interpretation by the actual patient, so it is

 

 19   something that is very difficult to evaluate, and

 

 20   that is why I put the continuum, because it is all

 

 21   there and you really shouldn't take a patient

 

 22   population based on angina or based on shortness of

 

 23   breath.

 

 24             I think you have got to bring both of

 

 25   these things together to understand they are sort

 

                                                                75

 

  1   of in the spectrum of a similar underlying

 

  2   pathophysiologic process.

 

  3              DR. SIMONS:  I would like to come back to

 

  4   the issues of the differences among the patients

 

  5   having these kind of therapies.  We have learned

 

  6   from a number of trials of growth factor therapies

 

  7   that there is a very large difference in how the

 

  8   patients respond.

 

  9             This issue that there are different

 

 10   subgroups that we are not defining is fairly

 

 11   critical to the field.  You mentioned one or two

 

 12   biomarkers, but there seemed to be a general

 

 13   association of markers as opposed to really

 

 14   identifying which patients respond in which manner.

 

 15             What would you suggest as a way of trying

 

 16   to sort of stratify these patient groups?  Not

 

 17   suggest ejection fraction, that is probably in a

 

 18   way sort of crude measure, but in terms of

 

 19   biological responses.

 

 20             DR. PERIN:  If we look at the trials of

 

 21   devices, I think that probably a common way to look

 

 22   at these patients is exercise capacity.

 

 23             I think that probably is one of the

 

 24   unifying parameters that we cannot only use at

 

 25   entry, but also you are able to follow as a patient

 

                                                                76

 

  1   goes along, and if he has a response to therapy, he

 

  2   will have a positive response in terms of what he

 

  3   is able to do in terms of function.

 

  4             That has a very practical translation into

 

  5   quality of life and people feeling better.  I would

 

  6   say in a broad sense, that exercise capacity, peak

 

  7   oxygen consumption might be something that I might

 

  8   consider an important thing to follow in these

 

  9   patients, and not just ejection fraction, which is

 

 10   dependent on a lot of things, how much loading the

 

 11   ventricle has that day, the amount of mitral

 

 12   regurgitation, et cetera, so there is a lot of

 

 13   things that will make that extremely variable.

 

 14             DR. RAO:  As an extension of that, it's a

 

 15   very general question.  Is there any problem with

 

 16   many of these studies which are in high-risk

 

 17   patients enrolling people for the placebo arm of

 

 18   the trial?  Not in cell therapy, but maybe when you

 

 19   do devices or you do assists, has this been

 

 20   historically a problem for the cardiovascular

 

 21   field?

 

 22             DR. PERIN:  Well, it has been done as you

 

 23   can see, so I have showed you a bunch of studies

 

 24   where it has been done, and it can be done.

 

 25             Personally, the way I like to see it is I

 

                                                                77

 

  1   want to offer patients that get in the placebo arm

 

  2   some kind of a treatment, so in our future upcoming

 

  3   study, what I am going to do is I will tell a

 

  4   patient you are going to get randomized to maybe

 

  5   not getting treatment, but if you don't get that

 

  6   treatment at an X period of time, six months, you

 

  7   will cross over to get the treatment.

 

  8             I think that is a humane way of doing it,

 

  9   in which these patients are very ill and desperate

 

 10   to get something to help, so again, if you can

 

 11   cross over, sometimes these placebo patients at

 

 12   some point after you have achieved your assessment,

 

 13   then that makes it a more palatable or fair way to

 

 14   do things maybe.

 

 15             DR. RAO:  Dr. Cunningham.

 

 16             DR. CUNNINGHAM:  I just wonder, in your

 

 17   data, if  you see any difference by either

 

 18   socioeconomic status or by gender, or by any way of

 

 19   culture, dividing populations, whether it would be

 

 20   race or ethnicity or any other factor like that?

 

 21             DR. PERIN:  You mean in our own--

 

 22             DR. CUNNINGHAM:  Yes, reading the JACC

 

 23   data, was there anything by gender, for instance,

 

 24   or by subpopulation?

 

 25             DR. PERIN:  Females, there are some

 

                                                                78

 

  1   differences in the female population in which there

 

  2   are some differences. There is the catch-up

 

  3   phenomenon in the end, but socioeconomic

 

  4   differences, I am not aware that it would have an

 

  5   impact on that, as well, but maybe gender

 

  6   differences, yes.

 

  7             DR. RAO:  One question to sort of follow

 

  8   on Dr. Simons' question, in at least the way I

 

  9   understood it, it is really kind of difficult to

 

 10   stratify patients or to extrapolate from one class

 

 11   of patients to the other.  Historically, that has

 

 12   always been a problem.

 

 13             Again, it's a general feeling when one

 

 14   conducts studies in the cardiovascular field, is

 

 15   there some consensus  that everybody says that,

 

 16   well, if you measure by ejection fraction, and we

 

 17   take patients, which is what it seemed like a lot

 

 18   of studies have done, that that is a reasonable

 

 19   criteria that you can extrapolate from one

 

 20   classification of that kind to the next, or one

 

 21   cannot?  Just as a general statement.

 

 22             DR. PERIN:   It has been done, and it is a

 

 23   general way of separating--there is definitely a

 

 24   correlation with your ejection fraction and your

 

 25   survival, so it is probably not the most refined

 

                                                                79

 

  1   way of dividing patients, and it depends where you

 

  2   make the cutoff, so if you make a fairly high

 

  3   cutoff, let's say, patients that had ejection

 

  4   fraction less than 40 percent, then, you are

 

  5   including most of the population of patients that

 

  6   have heart failure, so it's a general way to divide

 

  7   things.

 

  8             If you start decreasing that number of

 

  9   that cutoff, then, you are really selecting out

 

 10   more I think subpopulations we were talking about,

 

 11   maybe some different kind of subpopulations of

 

 12   patients with heart failure.

 

 13             DR. RAO:  Dr. Borer.

 

 14             DR. BORER:  Dr. Rao, a few minutes ago you

 

 15   made a point, and I would like to restate it in

 

 16   another way, because what Dr. Perin did, as I see

 

 17   it, is very well present an overview as an outline,

 

 18   was a scaffold upon which we can conduct subsequent

 

 19   more specific discussions.

 

 20             I think that right now we are getting into

 

 21   a series of questions that are way beyond the data

 

 22   that exist, and you couldn't expect Dr. Perin to

 

 23   respond to them in a meaningful way because the

 

 24   data don't exist.

 

 25                In specific response to your question,

 

                                                                80

 

  1   which was a very fundamental one, I think we are at

 

  2   a point now with this form of therapy where if we

 

  3   could define any group in which we saw a response

 

  4   which seemed credible, which was statistically

 

  5   valid, we would then have a series of hypotheses

 

  6   that would have been generated that would allow one

 

  7   to move further, but I think that is the level we

 

  8   are at.

 

  9             The idea of defining a general population

 

 10   in which to test therapy the way we do with drugs,

 

 11   we are not there yet, so I think the specific

 

 12   questions have to come a little later in this

 

 13   forum.

 

 14             DR. RAO:  I just wanted to get it clear to

 

 15   people that that was the case, but your point is

 

 16   very well taken.

 

 17             Dr. Neylan.

 

 18             DR. NEYLAN:  Thank you.

 

 19             That was a very nice clinical overview,

 

 20   and I wanted to ask you from your perspective as a

 

 21   clinician, there are obviously many parameters

 

 22   whose relief or improvement would be significant in

 

 23   terms of the lives of individual patients, and many

 

 24   of these could be utilized as endpoints for proof

 

 25   of concept.

 

                                                                81

 

  1             But ultimately, what do you believe is the

 

  2   most relevant clinical endpoint for defining

 

  3   registration criteria for this form of therapy, is

 

  4   it patient mortality or something else?

 

  5             DR. PERIN:  I don't know if we are going

 

  6   to be impacting patient mortality.  That is a very

 

  7   difficult question.  I would go back and what I had

 

  8   said earlier, and use an endpoint, I would use

 

  9   something like the LV02 as an endpoint.

 

 10             I think that is a little bit more

 

 11   palpable, and obviously, looking at mortality, this

 

 12   is such an initial incipient field in which we have

 

 13   barely treated any patient, so to think about

 

 14   looking at mortality, which involves a much larger

 

 15   number of patients, I think that is probably

 

 16   getting ahead of ourselves a little bit.

 

 17             We need to first verify if this is

 

 18   efficacious and if there is some objective

 

 19   improvement in these patients, and one of those

 

 20   objective ways of doing that would be something

 

 21   like exercise capacity, like I mentioned.

 

 22             DR. RAO:  Dr. Ruskin.

 

 23             DR. RUSKIN:  Just two quick comments on

 

 24   Dr. Perin's very nice presentation.

 

 25             One is that we have learned from drug and

 

                                                                82

 

  1   device trials that both ejection fraction and heart

 

  2   failure classification are critically important

 

  3   predictors, but that they are not necessarily fully

 

  4   interactive, that is, they are independent, so

 

  5   using both, I think in any classification with

 

  6   regard to these kinds of interventions would be

 

  7   critical because the outcomes are very, very

 

  8   different in Class III and IV even with the same

 

  9   EF.

 

 10             The other relates to a question that Dr.

 

 11   Rao raised about recruitment and controls.  I think

 

 12   that given the excitement in this area, but the

 

 13   unknown issues that have already been raised, doing

 

 14   trials that have adequate controls perhaps is more

 

 15   important here than anywhere else one can imagine

 

 16   given the severity of the illness that we are

 

 17   dealing with and the kinds of outcomes that Dr.

 

 18   Perin has described.

 

 19             As someone who recruits for device trials,

 

 20   though, I can tell you that it is not easy, and

 

 21   randomizing patients to acceptable controls in this

 

 22   kind of illness is going to be a huge challenge,

 

 23   but I think it is important for this group to

 

 24   emphasize that there is no place where this could

 

 25   be more important, otherwise, we will never get an

 

                                                                83

 

  1   answer, and I think that mortality ultimately will

 

  2   have to be a critical part of any trial that is

 

  3   done.

 

  4             DR. RAO:  Go ahead, Dr. Borer.

 

  5             DR. BORER:  I agree completely with Jeremy

 

  6   that controls are essential in this kind of

 

  7   research and really in any clinical research, but I

 

  8   think again to put this whole area in context, and

 

  9   in response to Dr. Neylan's point and question, we

 

 10   are at the point now of looking at physiological

 

 11   variables and what we would call in drug

 

 12   development "surrogates," to see whether cardiac

 

 13   performance, cardiac perfusion, this, that, and the

 

 14   other thing, is affected in one way or another, so

 

 15   that one could extrapolate to the point where it

 

 16   would be legitimate to define hypotheses about

 

 17   clinical outcome.

 

 18             We are not there yet, and the clinical

 

 19   outcome, just to put it in context from the drug

 

 20   world, is perfectly legitimate in the view of most

 

 21   people who deal with this area and these agents to

 

 22   think of a therapy as being approvable if it makes

 

 23   people feel better, but doesn't make them live

 

 24   longer.

 

 25             If it makes people feel better, even if it

 

                                                                84

 

  1   makes them live a little bit shorter, as long as

 

  2   you know how much shorter that is, and if it makes

 

  3   people live longer while not making them feel too

 

  4   much worse.

 

  5             I don't think we are at a point yet again

 

  6   to define what the outcomes variables should be.  I

 

  7   think we are at the point of defining physiological

 

  8   and pathophysiological surrogates, and that is what

 

  9   is being done in the studies to date, and then we

 

 10   can decide what the outcomes are, clinically

 

 11   important for registration.

 

 12             DR. RAO:  I guess that leads us to the

 

 13   fact that many of these things should be discussed

 

 14   tomorrow, just like  you pointed out.

 

 15             If there are no critical questions

 

 16   remaining, I will thank Dr. Perin.

 

 17             [Applause.]

 

 18             DR. RAO:  We are going to take a short

 

 19   break.

 

 20             [Break.]

 

 21             DR. RAO:  We are really extremely

 

 22   fortunate in having Dr. Menasch here to present

 

 23   his findings, and I look forward to a really

 

 24   interesting talk.

 

 25                Clinical Experience of Autologous

 

                                                                85

 

  1                     Myoblast Transplantation

 

  2             DR. MENASCHE:  Good morning.  First of

 

  3   all, I would like really to thank you for the

 

  4   privilege of this invitation and this unique

 

  5   opportunity of sharing some data on the clinical

 

  6   myoblast transplantation.

 

  7             What I would like to do in this talk is

 

  8   first to briefly touch on the preclinical data

 

  9   which have paved the way for these early clinical

 

 10   trials, and then, as requested by Dr. Grant, to

 

 11   focus on the various aspects of the clinical

 

 12   experience which has accumulated so far before

 

 13   drawing some perspectives which may have clinical

 

 14   relevance in the near future.

 

 15             Now, I think just to make things clear,

 

 16   that the basic assumption is fairly

 

 17   straightforward, and the objective of this therapy

 

 18   is really to try to repopulate areas of dead

 

 19   myocardium with new contractile cells with the hope

 

 20   that these areas can regain some function, and

 

 21   given the close relationship between function and

 

 22   survival, which has been already mentioned, the

 

 23   ultimate hope is obviously that it can have a

 

 24   significant impact on clinical outcomes.

 

 25             The reason why we initially started with

 

                                                                86

 

  1   the skeletal myoblasts are actually listed here.

 

  2   These cells are not really stem cells, they are

 

  3   better termed precursor cells for muscular fibers

 

  4   in that they are very committed to their skeletal

 

  5   muscle phenotype as you will see.

 

  6              The first advantage of the myoblasts is

 

  7   that they can be very easily retrieved from the

 

  8   patient himself, thus overcoming any problem

 

  9   associated with rejection and immunosuppressive

 

 10   therapies.

 

 11             These cells feature a very great expansion

 

 12   potential which is important given the relationship

 

 13   which exists between the number of cells which are

 

 14   injected and the ultimate functional outcome.

 

 15              As I have just said, they are pretty well

 

 16   committed to their myogenic lineage, and the risk

 

 17   of tumor development is virtually negligible.

 

 18   Finally, they are pretty resistant to ischemia, and

 

 19   although unfortunately, many of them die shortly

 

 20   after the injections, fortunately, some of them

 

 21   will survive and may positively affect function.

 

 22             So, this is type of animal model which has

 

 23   been used initially in rodents.  You see here the

 

 24   heart and the needle injecting the cells.  I just

 

 25   would like to mention that it took us seven years,

 

                                                                87

 

  1   seven years of preclinical work before I did

 

  2   operate on the first patient June 15, 2000.

 

  3             During the seven years, we moved from the

 

  4   rodent models to the large animal models, which I

 

  5   think is absolutely necessary before arriving to

 

  6   clinical trials.

 

  7             Just to summarize the bulk of this data,

 

  8   we can say, number one, that when you inject

 

  9   skeletal myoblasts into an infarcted area, they

 

 10   retain the possibility of differentiating into

 

 11   typical myotubes.  Here is a typical myotube,

 

 12   elongated structure, and this is a sheep heart and

 

 13   this is a human heart.

 

 14             This is an autopsy specimen.  One patient

 

 15   of our Phase I trial died 18 months after his

 

 16   surgery from stroke, and we had permission for the

 

 17   autopsy.  You will appreciate the striking

 

 18   similarity of these two slides.  Here you find in

 

 19   this human heart, a typical myotube embedded in

 

 20   scar tissue.

 

 21             At closer magnification, you can

 

 22   appreciate the typical cross-striations, and I

 

 23   think two observations are important to be made at

 

 24   this point.  Number one, these cells really remain

 

 25   committed to their skeletal muscle phenotype. In

 

                                                                88

 

  1   other words, there is virtually no evidence that

 

  2   they can ever turn to cardiomyocytes.  They will

 

  3   not become cardiac cells.

 

  4             Number two, they remain electrically

 

  5   insulated from the surrounding myocardium, which

 

  6   obviously raises major mechanistic questions

 

  7   regarding the underlying mechanisms by which they

 

  8   can improve function, but the fact is that there is

 

  9   no real evidence that they develop connections with

 

 10   the neighboring cardiomyocytes.

 

 11             Nevertheless, when you subject them to

 

 12   strong depolarizing currents, they show excitable

 

 13   properties, and you see here, this is a fluorescent

 

 14   myotube which has been grafted in a myocardial

 

 15   scar.  This is an in vivo study and definitely they

 

 16   can respond to currents by generating action

 

 17   potentials followed by contractions.

 

 18             This translates into an improvement in

 

 19   function, both regional function here in the sheep

 

 20   model, and global function, the LV ejection

 

 21   fraction.  This improvement, as you can see, seems

 

 22   to be sustained over time until one year in our rat

 

 23   studies, and basically, these kinds of observations

 

 24   have been made by several other investigators

 

 25   already past 10 years.

 

                                                                89

 

  1             So, there is a fairly good consistency

 

  2   showing that these myoblasts can, to some extent,

 

  3   improve function at least in animal models, and

 

  4   obviously, the gap with the humans is a wide one.

 

  5             So, if we now move to the clinical

 

  6   experience, so far there are 44 patients who have

 

  7   been included in early Phase I trials, and 34

 

  8   patients currently included in our ongoing

 

  9   randomized, multi-centered Phase II study .

 

 10             This list is by far not exhaustive.  I

 

 11   have not tabulated anecdotal case or me-too cases.

 

 12   I have just kept those studies which have been

 

 13   published in peer-reviewed journals.

 

 14             Basically, the inclusion criteria have

 

 15   been fairly straightforward across all these

 

 16   studies.  Patients with low ejection fractions,

 

 17   usually below 35 percent, patients with a history

 

 18   of myocardial infarct, and obviously, patients

 

 19   requiring concomitant coronary bypass surgery since

 

 20   for ethical reasons, it is difficult to open the

 

 21   chest just for injecting a product we don't really

 

 22   know whether it is effective or not.

 

 23             If we try to summarize the main results,

 

 24   we can say, number one, that multiple epicardial

 

 25   injections look to be safe.  I have never seen any

 

                                                                90

 

  1   bleeding from the needle holes, and overall, this

 

  2   experience has been shared by the other surgeons

 

  3   who have practiced the operation.

 

  4             Number two, it is possible--and we will

 

  5   come back on that--that the procedure increases the

 

  6   risk of arrhythmia postoperatively, at least in the

 

  7   early post-op period.

 

  8             Number three, I will be extremely careful

 

  9   and cautious about that, there are some data

 

 10   suggesting that maybe function can improve, but it

 

 11   is clear that until we have the results of the

 

 12   ongoing randomized, placebo-controlled study, we

 

 13   cannot make any meaningful conclusion.

 

 14             This is the list of the studies and of the

 

 15   patients.  I have just added the last one a few

 

 16   days ago. Professor Siminiak presented at the

 

 17   American College of Cardiology another series of 10

 

 18   patients who got the cells through a percutaneous

 

 19   catheter using the coronary sinus route.  I will

 

 20   come back on that catheter in a few minutes, but I

 

 21   will rather concentrate on the surgical

 

 22   implantations listed here.

 

 23             Dr. Smits also injected cells through a

 

 24   catheter using the interventricular approach

 

 25   similar to the one alluded to by Dr. Perin.

 

                                                                91

 

  1              This goes back to the inclusion criteria

 

  2   which have previously been mentioned.  I think it

 

  3   is important to look at all words, because as you

 

  4   will see, differences in definition may really be

 

  5   confounders in the interpretation of the results.

 

  6             It is important to look at akinetic areas

 

  7   that is really dead myocardium, not simply

 

  8   ipokinetic or dyskinetic, really akinetic

 

  9   myocardium, which are not amenable to

 

 10   revascularization and obviously, it is also

 

 11   important that the bypass surgery be done in other

 

 12   areas.

 

 13             For example, you will see that in one

 

 14   study, the area which was transplanted with cells

 

 15   was also revascularized, so when the authors

 

 16   conclude that cell therapy improves function, it is

 

 17   clearly meaningless since the same area has got

 

 18   simultaneous revascularization.

 

 19             For those of you who are not familiar with

 

 20   the procedure, I just would like briefly to show

 

 21   you this three-step operation.  It starts with a

 

 22   muscular biopsy.  We take it at the thigh.  It is a

 

 23   very simple procedure under local anesthesia.

 

 24             We remove a chunk of muscle, which is then

 

 25   cut into small pieces, put in this sheeping medium

 

                                                                92

 

  1   and sent to the cell culture lab where a multiple

 

  2   tri-cell factory is being designed to allow for

 

  3   large-scale cell production.

 

  4             Then, there are regular morphological

 

  5   controls. Obviously, the key point is to inject the

 

  6   cells before they reach confluence.  What you would

 

  7   like to do is that confluence occurs in vivo

 

  8   following the engraftment, not before, so it is

 

  9   important to check the morphological state of the

 

 10   cells on a regular basis.

 

 11             This is how human myoblasts look like

 

 12   during the cell culture process, and this is how

 

 13   the cells look like when they are back in the

 

 14   operating room.

 

 15             Then, with the curved needle, we inject

 

 16   the cells all across the infarcted area including

 

 17   the borders.  It's a time-consuming, I would say

 

 18   10, 12, 15 minute procedure, rather tedious and

 

 19   boring procedure, by the way, where you have to

 

 20   mentally construct the grids and then go with the

 

 21   needle from side to side, so we are working on the

 

 22   multiple shot device, but it is more tricky than we

 

 23   initially thought.

 

 24             So, right now we have the requirement for

 

 25   these multiple injections all across.  This is

 

                                                                93

 

  1   another view of the injections.

 

  2             So, if we start by feasibility, I think it

 

  3   is quite well established that this technique is

 

  4   perfectly feasible.  In other words, it does

 

  5   demonstrate that provided  you have the appropriate

 

  6   techniques, you can take a small piece of muscle

 

  7   which contains, say, 3- 4 million skeletal

 

  8   myoblasts initially and expand it over two to three

 

  9   weeks until approximately 1 billion cells.

 

 10             These are the results of our cultures

 

 11   during the Phase I trial, during which the target

 

 12   numbers which have been prespecified have

 

 13   consistently been obtained and even overshoot it.

 

 14             You will note that you can get up to 90

 

 15   percent of skeletal myoblasts in that--and this is

 

 16   an important point--you really end up with a pretty

 

 17   well defined cell therapy product.  You really know

 

 18   what you are injecting.

 

 19             Importantly, what we have seen is that

 

 20   heart failure does not prevent skeletal myoblasts

 

 21   to differentiate  into myotubes, and this was a

 

 22   question because when we did preclinical rounds, I

 

 23   got pieces of tissue from orthopedic colleagues,

 

 24   but often these patient were young, and the

 

 25   question was are the myoblasts from this Class

 

                                                                94

 

  1   III/IV heart failure patients going to

 

  2   differentiate normally, and the answer is yes, so

 

  3   far we have had no failure.

 

  4             The only thing is that it may take a

 

  5   little bit more time for some patients until we get

 

  6   the target number of cells, but at the end of the

 

  7   day, it has always been possible to achieve the

 

  8   prespecified target number of cells in myoblasts.

 

  9             What about safety now?  These are the

 

 10   different adverse events we were concerned with by

 

 11   the time we started the trial, and fortunately, I

 

 12   must say that none of them has occurred except--and

 

 13   we are going to discuss that--possibly the

 

 14   arrhythmias, but it is important to emphasize that,

 

 15   for example, there was never any particular

 

 16   bleeding from these multiple puncture sites.

 

 17             There was no unusual complication in the

 

 18   postoperative course of these patients, and when

 

 19   the cells were injected in newt immunocompromised

 

 20   mice, there was never any evidence for tumor

 

 21   formation.

 

 22             Obviously, before we started the study, we

 

 23   had to go through a lot of regulatory constraints,

 

 24   indeed, what I did is to discuss with the French

 

 25   FDA and ask them what was approved or not, and the

 

                                                                95

 

  1   game was not so easy because as previously

 

  2   mentioned, there was no precedent.

 

  3             So, they told us, well, this is what you

 

  4   are allowed to do.  This is the kind of culture

 

  5   medium, ancillary product additives which are

 

  6   permitted for human use, so we immediately from the

 

  7   onset designed our cell culture in accordance to

 

  8   the prespecified instructions, and obviously, it

 

  9   was timesaving because when we came back with the

 

 10   process, there was nothing else than to accept it.

 

 11             Well, what about the V-tachs?  In the

 

 12   initial series we had 4 patients with sustained

 

 13   episodes of ventricular tachycardia.

 

 14             All of them occurred during the early

 

 15   post-op period, the early three first week,

 

 16   postoperative weeks, and there was virtually no

 

 17   recurrence later on because these patients had a

 

 18   defibrillator put on and only one of them

 

 19   experienced firing of the defibrillator one year

 

 20   later, so it really appears to be a relatively

 

 21   early post-op event.

 

 22             Now, there are different mechanisms which

 

 23   could account for these arrhythmias, in particular,

 

 24   the differences in electrical membrane properties

 

 25   between the grafted cells and the neighboring

 

                                                                96

 

  1   cardiomyocytes.  Obviously, other mechanisms can

 

  2   also be considered, but we really favor the first

 

  3   one because we did an EP study in which we looked

 

  4   at the different membrane properties of the cells.

 

  5             Here, you see a typical action potential

 

  6   of a muscular fiber and here of a cardiomyocyte.

 

  7   Now, if you graft skeletal myoblasts back into a

 

  8   muscle, these cells retain a typical skeletal

 

  9   muscle phenotype, and this is also true for

 

 10   myotubes which grow in culture.

 

 11             The question is how does it look like when

 

 12   you graft the skeletal myoblasts into the heart.

 

 13   Well, definitely it remains very similar to what it

 

 14   was initially and different from the action

 

 15   potential of the cardiomyocyte.

 

 16             If you expressed it graphically, you would

 

 17   see that the action potential duration is quite

 

 18   different between the cardiomyocyte and the

 

 19   myotube, and this heterogeneity might account for

 

 20   some of these arrhythmias.

 

 21             Now, having said that, the picture is

 

 22   probably more complex and the reason, as you know,

 

 23   and it has been mentioned by Dr. Perin in his talk,

 

 24   is that heart failure by itself predisposes

 

 25   patients to arrhythmias.

 

                                                                97

 

  1             So, I think that as long as we don't have

 

  2   the results of the randomized trial in which all

 

  3   patients have been instrumented with a

 

  4   defibrillator, it will be difficult to conclusively

 

  5   establish a causal relationship between grafting of

 

  6   cells and the occurrence of arrhythmia.

 

  7             I can also tell you that we currently have

 

  8   randomized 34 patients in the Phase II trial and

 

  9   the incidence of arrhythmia has been strikingly

 

 10   low, much lower than in the initial study we had

 

 11   done, so things are probably less clear than they

 

 12   were initially, and once again we have to wait for

 

 13   the results of the randomized trial before we can

 

 14   definitely say yes, there is no relationship

 

 15   between myoblast transplantation and arrhythmia.

 

 16             Anyway, these patients or most of them

 

 17   would require at one point a defibrillator, so it

 

 18   was not a big issue for us to implant those

 

 19   defibrillators in all the Phase II patients.

 

 20             Now, what about efficacy?  Now, we have to

 

 21   be extremely careful in the interpretation of the

 

 22   results which are presented because of the

 

 23   multiplicity of the confounding factors.

 

 24             The culture conditions, for example, the

 

 25   Spanish group has used a culture medium which

 

                                                                98

 

  1   contains the patient's own serum, and the

 

  2   conclusion is we had no arrhythmia, so if you use

 

  3   the patient's own serum instead of fetal calf

 

  4   serum, you prevent arrhythmia.

 

  5             I think it is really a simplistic

 

  6   conclusion based on 12 patients, but it can

 

  7   introduce an additional bias.  There is currently

 

  8   no evidence that fetal calf serum is really

 

  9   responsible for the arrhythmias.

 

 10             Dosing has been extremely different and

 

 11   variable from one study to the other, as well as

 

 12   the kinetics of the  grafted area.

 

 13             Once again, any kinetic area is different

 

 14   from a dyskinetic area, which features a

 

 15   paradoxical motion, and, for example, in the U.S.

 

 16   trial, some patients were included who had

 

 17   hypokinesia, which we know can improve just because

 

 18   of the revascularization even if revascularization

 

 19   is not targeted at this particular area.

 

 20             The same for bypasses.  In the Spanish

 

 21   study, for example, the cell grafted areas were

 

 22   also bypassed, which makes the interpretation of

 

 23   results impossible.

 

 24             Type of surgery has also been different.

 

 25   In the U.S. study, for example, some patients had

 

                                                                99

 

  1   additional reconstructions of the left ventricle in

 

  2   addition to the bypass surgery, which make things

 

  3   still more complicated.

 

  4             Finally, the method of outcome assessment,

 

  5   in some studies, the assessment has been

 

  6   centralized at one side, in others, each center has

 

  7   made its own assessment, which obviously makes big

 

  8   differences.

 

  9             This is just to illustrate the variability

 

 10   in the number of cells which have been injected.  I

 

 11   don't have the figures for the initial surgical

 

 12   study from Professor Siminiak, but as you can see,

 

 13   there is a wide variability.

 

 14             The U.S. study of Dr. Dib was, as you

 

 15   know, was a dose escalating study accounting for

 

 16   this variability in the numbers.  Dosing is

 

 17   probably important.  This is one study among others

 

 18   showing that there seems to be a tight relationship

 

 19   between the number of injected cells and the

 

 20   functional outcomes.

 

 21             This is the reason why, in our early Phase

 

 22   I trial, we have targeted a high number of cells,

 

 23   800 million. In the Phase II, we have two arms with

 

 24   two different doses of cells, but the number

 

 25   probably makes a big difference given the high rate

 

                                                               100

 

  1   of early cell death.

 

  2             The characteristics of the grafted

 

  3   segments, as I previously mentioned, have also been

 

  4   different from one study to the other, as well as

 

  5   the method for assessing viability, usually,

 

  6   dobutamine echocardiography, occasionally MRI or

 

  7   PET scan.

 

  8              Same variability in the characteristics

 

  9   of injections, but you see that you can go up to

 

 10   almost 60 injections without any concern related to

 

 11   bleeding, and obviously, the number of injections

 

 12   depends on the extent of the area of infarction.

 

 13             It is also important to look at the cell

 

 14   concentration.  We extensively studied that before

 

 15   I started doing patients.  You have to find a

 

 16   tradeoff because if you use a large needle, then,

 

 17   you can have large holes and some bleeding

 

 18   problems.

 

 19             If you use a too small needle, you will

 

 20   eliminate the bleeding problems, but the cells may

 

 21   be packed and damaged through their passage, so we

 

 22   ended with a 27-gauge needle which gave an

 

 23   acceptable rate of cell viability.

 

 24             The concentration of cells is important,

 

 25   and probably still more important when you are

 

                                                               101

 

  1   using a long catheter.  We are using a short needle

 

  2   with directly the serum hooked to the needle, but

 

  3   if you are using a long catheter, concentration may

 

  4   make a big difference.

 

  5             Finally, revascularization is occasionally

 

  6   being done in the same area as the area where cells

 

  7   were put in, which completely confuses the results.

 

  8             This is, for example, the Spanish study,

 

  9   what you see is that, what they call the untreated

 

 10   segments, that it is segments which had just

 

 11   bypassed, the wall motion score went from 1.2 to

 

 12   1.1 and 1, but really, this is almost normal

 

 13   motion, so obviously, it makes it easier to

 

 14   demonstrate that in the other segments which have

 

 15   bypass surgery and cells, the improvement was

 

 16   greater.

 

 17             This is a summary of our data from the

 

 18   Phase I trial.  We had an improvement in the

 

 19   functional status and an increase in ejection

 

 20   fraction.  These results are meaningless because

 

 21   these patients had associated bypass surgery.

 

 22             So, we rather looked at the number of

 

 23   scarred segments, and I remind you these were

 

 24   akinetic segments without viability on dobutaminic

 

 25   echocardiography without any possibility for

 

                                                               102

 

  1   revascularization.  So, we looked at the changes in

 

  2   the contractions of these segments which have been

 

  3   grafted with cells.

 

  4             So, initially, obviously, there was no

 

  5   motion since it was one of the inclusion criteria,

 

  6   and afterwards we had, at two different time

 

  7   points, approximately 60 percent of segments

 

  8   regaining some function.

 

  9             I am not saying that these segments were

 

 10   normally contracting, they were not.  There was a

 

 11   slight and modest improvement.  This was a blinded

 

 12   assessment, in other words, we blinded the dates of

 

 13   the echo tapes and asked independent

 

 14   echocardiographers to review them and to grade

 

 15   them.  There was a modest improvement, not normal

 

 16   contraction, but it was sufficient to push us to

 

 17   move forward to the Phase II study.

 

 18             I just show you a couple of examples.

 

 19   This is a flat exterior wall, no motion at all, and

 

 20   this is the same wall with the systolic thickening

 

 21   following myoblast transplantation.  This is the

 

 22   MRI study which does not project on the screen.  I

 

 23   have it on the computer, but not on the screen.

 

 24             You see here the interior infarct which

 

 25   has been grafted, and you can appreciate an

 

                                                               103

 

  1   improvement in wall motion in the postoperative

 

  2   period.  This is an exterior infarct.  You see the

 

  3   thin wall here, which has been grafted, and this is

 

  4   the post-op pattern with a thickening of the wall.

 

  5             I add intentionally that these patients

 

  6   also had bypasses in the left system.  I don't like

 

  7   the slides where you see pre-transplantation,

 

  8   post-transplantation, just omitting that in

 

  9   addition, there was either bypass surgery or

 

 10   balloon angioplasty.

 

 11             This is another example of an interior

 

 12   infarct pre-transplantation and bypass to the

 

 13   posterior descending coronary artery and the

 

 14   post-op, with an improvement in the wall motion.

 

 15             So, now, can it be due to the

 

 16   revascularization of the PDA?  It is unlikely, but

 

 17   it cannot be eliminated.

 

 18             So, basically, this is the design of the

 

 19   MAGIC, the Phase II trial which has been initiated

 

 20   now in Europe, in different countries in Europe.

 

 21   It is targeted to include 300 patients in different

 

 22   countries, and to emphasize what Dr. Ruskin was

 

 23   mentioning earlier, it is a placebo-controlled

 

 24   study.  In other words, patients following

 

 25   randomization have a muscular biopsy and they have

 

                                                               104

 

  1   eventually injection of a placebo solution in

 

  2   addition to their bypass surgery.

 

  3             There are three arms, one control and two

 

  4   treated groups, one having 400 million, the other

 

  5   having 800 million cells.  The production of cells,

 

  6   and this is probably important, has been

 

  7   centralized in two sites, one in Paris and one in

 

  8   Boston, and it is exactly the same technology which

 

  9   is used in the two sites.

 

 10             The primary endpoint is the improvement in

 

 11   the contractility of the segments which have been

 

 12   grafted with cells in the core lab and in a blinded

 

 13   fashion.  In addition to that, we are obviously

 

 14   looking at major adverse cardiovascular events at

 

 15   the one-year follow-up time.

 

 16             I would like to move on now before

 

 17   finishing to some clinically relevant perspectives

 

 18   which may have really clinical implications in the

 

 19   near future.

 

 20             First of all, so far we have been talking

 

 21   primarily of ischemia cardiomyopathy, but as

 

 22   mentioned by Dr. Perin, there are other causes of

 

 23   heart failure in particular non-ischemic, globally

 

 24   dilated cardiomyopathy.

 

 25             So, we have been interested in assessing

 

                                                               105

 

  1   myoblast transplantation in this particular

 

  2   context, and use a particular genetic strain of

 

  3   hamsters which develop a non-ischemic dilated

 

  4   cardiomyopathy, and randomize the animals to

 

  5   receive either autologous skeletal myoblasts,

 

  6   because phenotypically, these myoblasts are free

 

  7   from the disease, or culture medium.

 

  8             To make a long story short, you see that

 

  9   there is a definite improvement in function which

 

 10   correlates with a major engraftment of cells in

 

 11   this non-ischemic myocardium. I think it just

 

 12   brings another piece of evidence that maybe

 

 13   something good is occurring.

 

 14             The second problem is cell death.

 

 15   Regardless of the cell type, cell death is

 

 16   extremely high, 80, 90 percent of cells are dying

 

 17   shortly after the injections for a variety of

 

 18   causes, in particular, apoptosis, but also

 

 19   ischemia.  It makes sense since we are injecting

 

 20   cells in scar areas which receive very little

 

 21   vascularization.  So, even if myoblasts are fairly

 

 22   resistant, they die nevertheless.

 

 23             So, now there are several studies

 

 24   suggesting that the co-induction of angiogenesis

 

 25   may be an effective means of improving survival of

 

                                                               106

 

  1   the cells, and ultimately, of improving function of

 

  2   the animals.

 

  3             This is a study comparing transplantation

 

  4   of fetal cardiomyocytes, injection of fibroblast

 

  5   growth factor, or a combination of both.  As you

 

  6   can see, function is improved when you combine the

 

  7   two therapies.

 

  8             Recently, we have duplicated this study

 

  9   except that we used myoblasts and another growth

 

 10   factor, and we found exactly similar results.

 

 11             So, there are different ways of inducing

 

 12   angiogenesis, and I know Dr. Epstein is going to

 

 13   discuss that, but the point I wanted to make, this

 

 14   is, you know, the difference in cell survival

 

 15   between myoblasts alone and myoblasts plus an

 

 16   angiogenic growth factor.

 

 17             The point I would like to make is that

 

 18   probably in the future, you will have to deal with

 

 19   proposal of studies trying to combine cell

 

 20   transplantation with some form of angiogenesis just

 

 21   to optimize cell survival and potentiate the

 

 22   benefits of the intervention.

 

 23             A third point regards cycling.  This is

 

 24   the muscular biopsy of the patient who died.  I

 

 25   previously talked about this patient who died from

 

                                                               107

 

  1   a stroke.  Initially, in this biopsy, and this is

 

  2   not unexpected, you find fast skeletal myosin and

 

  3   slow type myosin.  You don't find fibers, virtually

 

  4   no fibers co-expressing fast and slow.

 

  5             When we looked at the heart of this dead

 

  6   patient, we found approximately 30 percent of cells

 

  7   co-expressing fast and slow myosin, which means

 

  8   that although once again these myotubes remain

 

  9   myotubes and do not turn to cardiac cells, it seems

 

 10   that some of them may incur some phenotypic changes

 

 11   in response to their new myocardial environment and

 

 12   start expressing slow myosin, which as you know is

 

 13   a fatigue-resistant myosin.

 

 14             So, this is important and should be put in

 

 15   parallel with this study showing that if you

 

 16   co-culture myoblasts in cardiac cells,

 

 17   cardiomyocytes, this is the green myoblast, this is

 

 18   an antibody against a cardiac troponin and against

 

 19   another cardiac marker, some of the myoblasts, as

 

 20   you can appreciate here, will express some cardiac

 

 21   markers.

 

 22             Now, what is shown here is that if you

 

 23   stop the beating of the co-culture fetal cells,

 

 24   there is no myoblasts which can acquire cardiac

 

 25   cell characteristics.  Conversely, if you subject

 

                                                               108

 

  1   the preparation to a cyclic stretch, then, the

 

  2   stretch makes some of these myoblasts able to

 

  3   express the cardiac markers.

 

  4             In other words, it is quite possible that

 

  5   in vivo, the cyclic contraction of the neighboring

 

  6   cardiomyocytes leads to the expression of some

 

  7   cardiac markers and leaves the slow myosin in the

 

  8   grafted cells, and the practical implication could

 

  9   be that maybe combining cell transplantation with

 

 10   ventricular stimulation, resynchronization could

 

 11   actually improve the extent by which the grafted

 

 12   cells express slow myosin and become fatigue

 

 13   resistant.

 

 14             So, once again, because this is a

 

 15   clinically used modality, biventricular

 

 16   resynchronization, in the future, we may have to

 

 17   deal with studies trying to combine ventricular

 

 18   resynchronization with cell transplantation.

 

 19              Finally, a few words about the routes of

 

 20   delivery, I have talked about epicardial

 

 21   injections, we are also looking at the scaffolds,

 

 22   which are just put on top of the infarcted area.

 

 23   We are currently working on polyurethane, as well

 

 24   as collagen patches.  Obviously, it is less

 

 25   traumatic and maybe it could reduce a little bit

 

                                                               109

 

  1   the extent of cell death.  This is a pattern after

 

  2   a couple of weeks.

 

  3             Now, the catheters, I know that this issue

 

  4   will be discussed this afternoon.  I just want to

 

  5   say that from a surgical perspective, I am amazed

 

  6   by the fact that many clinical studies have been

 

  7   initiated in spite of the fact that we had few data

 

  8   on cell retention, functionality, cell viability is

 

  9   not the only issue.

 

 10             It is not because you see myoblasts, that

 

 11   they are going to turn into myotubes.  You really

 

 12   have also to assess the functionality, the ability

 

 13   for these cells to become myotubes, long-term

 

 14   engraftment, as well as the possible interactions

 

 15   between catheter materials and the cells.

 

 16             Most of the studies published so far in

 

 17   the preclinical setting have dealt with technical

 

 18   feasibility rather than functional efficacy, and

 

 19   the various routes have not really been compared.

 

 20             Having said that, we are very interested

 

 21   in the percutaneous routes, and in our group, we

 

 22   have interventional cardiologists working in that.

 

 23   I must say that we have been primarily interested

 

 24   by the transvenous cell injection through the

 

 25   coronary sinus, and this is the summary of the

 

                                                               110

 

  1   study which was presented last week at the ACC,

 

  2   which is a functional study that is a sheep model

 

  3   of myocardial infarction in which we injected cells

 

  4   through this catheter.

 

  5             You can appreciate that it allows a real

 

  6   delivery of the myoblasts, which turn into

 

  7   myotubes, and this correlated with a significant

 

  8   improvement in function.  So, this is a not a

 

  9   feasibility study, this is a true efficacy study,

 

 10   which is encouraging at least with regard to this

 

 11   particular catheter.

 

 12               So, these are maybe the challenges of

 

 13   the future, in the setting of bone marrow, the

 

 14   famous MAPS, the mesenchymous adult report in

 

 15   cells, which feature distant advantages, possible

 

 16   disadvantages.  We are currently working on the

 

 17   cells, cardiac progenitors, and I am sure Michael

 

 18   Schneider will have a lot of things to say about

 

 19   that.  Also, possible embryonic stem cells.

 

 20             What is also important now is to compare

 

 21   cells between them, and not exclusively with

 

 22   controls.  This is true, for example, with the bone

 

 23   marrow.

 

 24             I just would like to show you a recent

 

 25   study that we have done comparing skeletal

 

                                                               111

 

  1   myoblasts, CD133 progenitors or culture medium in a

 

  2   randomized study, and the result is that there is

 

  3   virtually no difference between the CD133 and the

 

  4   myoblasts in terms of function.

 

  5              If you look at histology, it is easy to

 

  6   find the myotube.  It is co-expressed, you know,

 

  7   specific markers like myosin heavy chain.  It has

 

  8   been extremely difficult to identify the CD133.  We

 

  9   have to rely on PCR to find some of them, which

 

 10   means that probably very few are still present

 

 11   after one month.

 

 12             So, I think it is important to compare the

 

 13   cells, and to some extent, given the amount of data

 

 14   which have accumulated over years, skeletal

 

 15   myoblasts may provide a sort of benchmark for

 

 16   testing other cell types.

 

 17             A similar study is being done in Doris

 

 18   Taylor's lab showing basically that there was no

 

 19   difference between skeletal myoblasts and

 

 20   mesenchymous cells.

 

 21             Once again, we are back to the question

 

 22   which was raised by Michael Schneider.  This is the

 

 23   setting of chronic heart failure, and in this

 

 24   particular setting, current evidence will rather

 

 25   favor skeletal myoblasts.

 

                                                               112

 

  1             This is a completely different setting

 

  2   from acute MI in which bone marrow cells seem to

 

  3   generate impressive results, but they are different

 

  4   patient populations, and it is quite possible that

 

  5   the acute stage of the MI, the bone marrow cells

 

  6   receive appropriate signals which allow them to

 

  7   improve function.  The setting may be quite

 

  8   different where you are dealing with heart failure

 

  9   patients and old scars for which apparently,

 

 10   skeletal myoblasts look more suitable for improving

 

 11   function.

 

 12             So, I just would like to close by two

 

 13   general slides summarizing a little bit what we

 

 14   have learned from our 10-year experience in the

 

 15   field.

 

 16             Regarding preclinical issues, it is clear

 

 17   that screening experiments have to be done in

 

 18   rodents, but I think it is critically important to

 

 19   validate that in large animal models before

 

 20   arriving to clinical trials.

 

 21             We have a good example of that with the

 

 22   combination of bone marrow cells and JCSF.  You are

 

 23   aware of the initial study by Orlig's group showing

 

 24   a regeneration of mouse myocardium by combining

 

 25   JCSF and bone marrow cell transplantation.

 

                                                               113

 

  1             This mouse study could now be duplicated

 

  2   by two independent groups including Orlig's group

 

  3   in primates, and then we have the Lancet paper last

 

  4   week showing that there was a higher rate of

 

  5   restenosis in patients receiving these two

 

  6   therapies.

 

  7             So, this jump from the mouse to the man

 

  8   without an intervening large animal model seems to

 

  9   be maybe a little questionable.

 

 10             It is also important that this preclinical

 

 11   study be designed just like clinical studies with

 

 12   appropriate controls and blinded assessment, but

 

 13   having said that, we must be aware that all these

 

 14   models have serious limitations at what point we

 

 15   are not able really to model the very complex

 

 16   situation of heart failure patients with a

 

 17   long-standing coronary artery disease.  A good

 

 18   example is that in our preclinical work, we have

 

 19   never seen any arrhythmia in any of the animals.

 

 20             Regarding clinical issues, it is important

 

 21   to have a well characterized cell therapy product.

 

 22   I am not sure that once the feasibility has been

 

 23   demonstrated in small pilot trials, it is necessary

 

 24   to multiply this 10-patient studies, because the

 

 25   amount of information that can be collected from

 

                                                               114

 

  1   these small studies is indeed limited once

 

  2   visibility has been established, and I think it is

 

  3   rather important to move on to larger clinical

 

  4   trials focusing in efficacy and safety, and

 

  5   allowing to draw more meaningful conclusions,

 

  6   safety, the arrhythmias with the myoblasts and

 

  7   possibly instant restenosis with the bone marrow,

 

  8   and efficacy obviously is left ventricular function

 

  9   and major inverse cardiovascular events.

 

 10             So, we are really now at very early stage,

 

 11   as you know, in the field.  We have some evidence

 

 12   that the myoblasts, among others, may improve

 

 13   function, but we still have a lot of basic

 

 14   questions to answer, and in the meantime, I don't

 

 15   think we can make any progress without the

 

 16   implementation of well-designed clinical trials

 

 17   more or less resembling those which have been

 

 18   designed for drugs with appropriate controls,

 

 19   randomization, blinded assessment, and so on,

 

 20   because this is the only way really to know whether

 

 21   hosts will be matched or not.

 

 22             I would like to acknowledge obviously all

 

 23   those who have participated in this endeavor with a

 

 24   special thanks for those who really did the work.

 

 25             Thank you very much.

 

                                                               115

 

  1             [Applause.]

 

  2             DR. RAO:  We will be open for questions.

 

  3             Go ahead, Dr. Borer.

 

  4                               Q&A

 

  5             DR. BORER:  First of all, Dr. Menasch, I

 

  6   have to tell you I think that was one of the most

 

  7   exciting talks I have heard in a long time.  That

 

  8   was really wonderful.

 

  9             I have some specific questions.  I will

 

 10   only give a couple of them, so that everybody else

 

 11   can talk, and then maybe ask a few more later.

 

 12             During your many years of preclinical

 

 13   studies, I am sure you made efforts to determine

 

 14   whether there were aspects of the preparation that

 

 15   could increase the plasticity of the myoblasts, so

 

 16   that they would manifest themselves more as

 

 17   cardiomyocytes than as myocytes.

 

 18             If you did, number one, did you find

 

 19   anything that altered the character, that increased

 

 20   plasticity, because if it did, that suggests that

 

 21   maybe the current preparation isn't the end of the

 

 22   line, maybe one could do better.

 

 23             With that in mind, you mentioned that you

 

 24   saw increased evidence of differentiation into

 

 25   cardiomyocytes or more cardiomyocyte

 

                                                               116

 

  1   characteristics in the beating setting.

 

  2             So, I wonder--I am sure you thought about

 

  3   it--but I wonder if you did culture any of the

 

  4   cells, rather than on flat plates, on flexor cell

 

  5   plates where periodic stress was applied in the

 

  6   culture phase, so that you could perhaps generate

 

  7   some of these cardiomyocyte characteristics before

 

  8   injection.

 

  9             That is one set of questions, and a second

 

 10   question I would like to just put in here, because

 

 11   it's a one-word answer, if these cells are

 

 12   electrically isolated, as you mentioned, how is it

 

 13   that they were caused to be in concert with the

 

 14   rest of the heart?

 

 15             DR. MENASCHE:  Regarding your first

 

 16   question, to be honest, we have not found any trick

 

 17   during the cell culture process which could really

 

 18   increase the transdifferentiation of these

 

 19   myoblasts into cardiomyocytes, and really, I don't

 

 20   think--I am thinking of the works of Chuck Murray,

 

 21   for example--I think no one has really shown that

 

 22   changes in the culture conditions could really make

 

 23   them turn into cardiomyocytes.

 

 24             The only evidence that can acquire some

 

 25   cardiac-like characteristics is this expression of

 

                                                               117

 

  1   slow type myosin.  We are currently exploring the

 

  2   possibility maybe of increasing the expression of

 

  3   these slow myosin isoform by implantation

 

  4   stimulation of the cells, but these are experiments

 

  5   which now are going to be done, and this was the

 

  6   reason why I was mentioning ventricular stimulation

 

  7   as a potential additive in the future to the

 

  8   clinical trials.

 

  9             But to summarize, no, we have not found

 

 10   any particular intervention, although maybe we have

 

 11   not found the right one, which could increase the

 

 12   proportion of cardiac-like skeletal myoblasts.

 

 13   Now, the mechanisms, I don't know; from scratch, I

 

 14   don't know.

 

 15             There are different possibilities.  One is

 

 16   a limitation of remodeling.  I am not sure if there

 

 17   is a predominant mechanism because in our patients,

 

 18   we have had some evidence of improved systolic

 

 19   function, but we have never seen a reduction in

 

 20   left ventricular diastolic dimensions.

 

 21             Another possibility is that GAB junctions

 

 22   are not the only ways for electrical impulses to

 

 23   travel across the heart, and as you have seen on

 

 24   the film, these cells retain excitable properties.

 

 25   In order words, if you excite them, they will

 

                                                               118

 

  1   contract.

 

  2             So, it is not completely impossible that

 

  3   in areas where physically, they are very close to

 

  4   the neighboring cardiomyocytes, they may be

 

  5   directly excited by electrotonic currents, and

 

  6   there is a third hypothesis we are currently

 

  7   exploring, and which is the paracrine hypothesis.

 

  8             It is quite possible, and it has been

 

  9   shown for bone marrow, for example, that these

 

 10   cells secrete various growth factors or cytokines,

 

 11   and so on, that can positively affect the function

 

 12   of the host cardiomyocytes.

 

 13             For example, in our studies we have found

 

 14   that myoblasts and myotubes from patients secrete

 

 15   very large amount of IGF-1, which has important

 

 16   effects on tissue regeneration.  So, maybe it has

 

 17   nothing to do with their countertype properties,

 

 18   but rather with the fact their behavior, small

 

 19   factors releasing good factors for the heart.

 

 20             So, we are currently playing with all

 

 21   these hypotheses, but I don't have any definite

 

 22   answer.

 

 23             DR. RAO:  So, it is pretty clear, like Dr.

 

 24   Schneider pointed out earlier, that mechanism is an

 

 25   issue that is still not clear.

 

                                                               119

 

  1             DR. MENASCHE:  No, it is not clear at all.

 

  2   The only thing is I don't think that you could

 

  3   infer from the lack of connexin 43 expression, that

 

  4   improvement in function is not possible.  I think

 

  5   both should be dissociated.

 

  6             DR. RAO:  Dr. Kurtzberg.

 

  7             DR. KURTZBERG:  You mentioned studies with

 

  8   bone marrow derived AC133 cells.  I wonder what the

 

  9   rationale behind the selection was and why you

 

 10   thought there would be an advantage to using

 

 11   selected cells over whole bone marrow.

 

 12             DR. MENASCHE:  The reason is that we first

 

 13   did a large animal study with whole bone marrow in

 

 14   the sheep model of myocardial infarction, once

 

 15   again, a chronic infarct.  So, we injected the

 

 16   whole bone marrow and we didn't find anything, no

 

 17   improvement in function, no limitation in

 

 18   remodeling, no evidence for transdifferentiation of

 

 19   cells.

 

 20             So, we said, well, maybe the whole bone

 

 21   marrow is not the appropriate medium for this

 

 22   particular setting, let's try to purify the cells,

 

 23   and we went to the CD133.  The results were

 

 24   slightly better in that.  There was some

 

 25   improvement in function compared with controls, but

 

                                                               120

 

  1   the improvement was not greater than that we got

 

  2   with the myoblast.

 

  3             Currently, we are comparing now myoblasts

 

  4   with AMAPCs, so we have tried to pick the different

 

  5   populations, in a stepwise approach, test all of

 

  6   them.

 

  7             DR. RAO:  Dr. Cannon.

 

  8             DR. CANNON:  Thank you for your talk, it

 

  9   was most interesting.  I am Richard Cannon from

 

 10   NHLBI.

 

 11             My question is in any of your preclinical

 

 12   animal work, did you ever inject your myoblast

 

 13   culture preparations or cell suspensions into the

 

 14   circulation to see where they might end up and what

 

 15   toxicity they might cause.

 

 16             This may not be an issue with an

 

 17   intra-operative injection into scar, but I would

 

 18   imagine with a catheter-based approach, it is

 

 19   conceivable that despite the operator's best

 

 20   efforts, some of these cells might be injected into

 

 21   systemic circulation.

 

 22             Do you have any data on where the cells

 

 23   end up, do they lodge in the brain or the kidneys,

 

 24   do they cause any toxicity or injury to other

 

 25   tissues?

 

                                                               121

 

  1             DR. MENASCHE:  Well, in the preclinical

 

  2   studies we have done, we have not found evidence

 

  3   for, first of all, all the injections were direct

 

  4   intramyocardial injections, so it may be difficult

 

  5   to find them in the brain or in the liver.

 

  6             We have not found them disseminated

 

  7   throughout the body, but I must say that maybe if

 

  8   we had done more extensive studies, autopsy studies

 

  9   of the brain or the lungs, or any other organ,

 

 10   maybe we could have found some of them.

 

 11             We have never injected intentionally the

 

 12   cells intravenously just to see what was happening.

 

 13   I don't believe it is a real issue because even

 

 14   when you are injecting them intraoperatively in

 

 15   humans, it is clear that some of them are escaping

 

 16   through the lymphatic system or in the venous

 

 17   system, and so far we have never seen any evidence

 

 18   for unexpected or unusual complications.

 

 19             But I agree with you that if you are

 

 20   expecting some leakage of the cells in the systemic

 

 21   circulation, this is probably a point that should

 

 22   be addressed more extensively than we have done.

 

 23             DR. RAO:  I have a practical question.

 

 24   Did you, when you looked at the cells, ever look at

 

 25   BRD incorporation to see whether cells continue to

 

                                                               122

 

  1   divide at any time?

 

  2             DR. MENASCHE:  Yes, absolutely, including

 

  3   in the human trial.  In the human trial, we always

 

  4   keep aliquot, initially, we kept aliquots of cells

 

  5   and just let them grow, and this is why we have

 

  6   been able to show that these cells were

 

  7   differentiating into myotubes including in heart

 

  8   failure patients.

 

  9             DR. RAO:  Did you ever take your samples,

 

 10   and look at freeze/thaw?  You know, you grow them

 

 11   in cell culture, can you freeze these cells and do

 

 12   they behave the same way when you send them to

 

 13   another site like you are planning in the Phase II

 

 14   trial, for example?

 

 15             DR. MENASCHE:  Absolutely, we have done

 

 16   that and we have validated that after thawing, they

 

 17   retain their ability to differentiate into

 

 18   myotubes.

 

 19             DR. RAO:  Have they been done in any

 

 20   transplant paradigm, or has it only been done by

 

 21   looking at they are forming myotubes in culture?

 

 22             DR. MENASCHE:  Both.  We have several

 

 23   preclinical studies in rats and in sheep, in which

 

 24   we have used cryopreserved and thawed cells with

 

 25   apparently functional outcomes similar to those we

 

                                                               123

 

  1   had with fresh primary myoblasts.

 

  2             This is the reason why actually we got

 

  3   permission to freeze them should they become

 

  4   necessary for logistical reasons in the Phase II.

 

  5             DR. BORER:  You mentioned that about 95

 

  6   percent of the cells that you inject have CD56

 

  7   characteristics.  That suggests that there is some

 

  8   alteration in some of the cells or perhaps a

 

  9   different cell line is growing in parallel, in the

 

 10   cultures that you are using, so I wonder, number

 

 11   one, how many passages do you use before

 

 12   administering the cells, and, number two, is the

 

 13   reproduction error rate increased with passage in

 

 14   any meaningful way, and does it make any

 

 15   difference?

 

 16             Obviously, a lot of these cells that you

 

 17   inject are nonviable.  When you inject them, they

 

 18   don't survive.  I don't know which ones are

 

 19   surviving and which ones aren't. But it seems to me

 

 20   that the number of passages employed may affect the

 

 21   ultimate outcome of the injection, and I wonder if

 

 22   you have some data on that from your preclinical

 

 23   work.

 

 24             DR. MENASCHE:  We used three to four

 

 25   passages, but it has been shown by Chuck Murray

 

                                                               124

 

  1   that if you multiply passaging, you may end up with

 

  2   a population of differentiation of effective cells,

 

  3   in which case you might end up with some unexpected

 

  4   overgrowth without any functional benefit.

 

  5             So, it is probably important not to

 

  6   multiply passaging too much.  With three to four

 

  7   passages, we have been able to reach the target

 

  8   numbers of cells, 400- or 800 million of cells.  We

 

  9   don't go beyond that.

 

 10             DR. TSIATIS:  In your randomized clinical

 

 11   trial, do you actually have formal stopping rules

 

 12   for either safety or efficacy, and, if so, what are

 

 13   they, or what is the general philosophy for

 

 14   monitoring?

 

 15             DR. MENASCHE:  It is primarily based on

 

 16   the judgments of the DMSB given the type of

 

 17   surgical population we are dealing with.  It is

 

 18   difficult to have stopping rules, just as you can

 

 19   have with drugs, for example, for each adverse

 

 20   event is reviewed by the DSMB, and based on that,

 

 21   they would decide whether the study has to be

 

 22   stopped or not.

 

 23             DR. RAO:  Dr. Neylan.

 

 24             DR. NEYLAN:  Thank you.

 

 25             I have another preclinical question.  I

 

                                                               125

 

  1   was wondering if you had an opportunity to compare

 

  2   the morphology and functionality of the myoblasts

 

  3   when these are injected either into the akinetic

 

  4   areas or perhaps into an area resected, and thus

 

  5   undergoing a normal reparative milieu, and whether

 

  6   perhaps under that milieu, there might be a

 

  7   different behavior of these cells or expression.

 

  8             DR. MENASCHE:  Really, basically, our

 

  9   model has been the model of, you know, coronary

 

 10   ligation creating myocardial infarction, so you

 

 11   really end up with an akinetic  scar.  I cannot

 

 12   answer this question.

 

 13             DR. NEYLAN:  You never had the chance to

 

 14   maybe resect that, maybe adhere to the natural

 

 15   surgical tendency of cutting things out.

 

 16             DR. MENASCHE:  I try to refrain from that.

 

 17   I have cardiologists as bodyguards, so it would

 

 18   just refrain you from doing that.

 

 19             DR. MULE:  You had mentioned that the vast

 

 20   majority of cells that are injected will die, and

 

 21   clearly, there is room for improvement with perhaps

 

 22   increasing angiogenesis, and so forth.

 

 23             About the kinetics of myotube formation in

 

 24   the ischemic areas, is it a dynamic process, in

 

 25   other words, once you inject the cells, myotubes

 

                                                               126

 

  1   will form over a given period of time, and then no

 

  2   more tubes will form, or additional tubes are

 

  3   generated over a prolonged period of time, and do

 

  4   those tubes, when they are formed, remain viable

 

  5   for the extension of the observation period?

 

  6             DR. MENASCHE:  It is difficult to tell

 

  7   you.  The kinetic studies indicate that although a

 

  8   substantial number of cells die, the remaining ones

 

  9   obviously proliferate in different shape over a

 

 10   period which seems to extend, say,  two to three

 

 11   weeks.

 

 12             At least in patients when we have seen

 

 13   improvements, we have never seen improvements

 

 14   before one month, and in animals, it is difficult

 

 15   to see any improvement before two weeks.

 

 16             Now, afterwards, the longest follow-up we

 

 17   have is 14 months in animals, but we cannot know

 

 18   whether the myotubes that we found at the end of

 

 19   the experiments were present since the beginning or

 

 20   whether they have been continuously regenerating.

 

 21             The interesting observation, however, I

 

 22   don't know whether it really answers your question,

 

 23   is that these myotubes harbor new myoblasts, so

 

 24   when you look at them with electron microscopy, you

 

 25   clearly see, on their basal lamina of these

 

                                                               127

 

  1   myotubes, newly formed myoblast cells, so they are

 

  2   able to regenerate their own pool of precursor

 

  3   cells.

 

  4             Now, whether these cells participate in

 

  5   the formation of new myotubes, I don't know.

 

  6             DR. RAO:  The last comment, Dr. Noguchi,

 

  7   and then we move on.

 

  8             DR. NOGUCHI:  I am sorry to have prolonged

 

  9   this, but it is just fascinating.  Of course, FDA

 

 10   always loves these controls, but to follow up on

 

 11   Dr. Mul's question, is it a question of liability,

 

 12   do the cells have to be alive, or you have a

 

 13   myotube has some structure, and then I just recall

 

 14   there is, in tumor biology, an old effect called

 

 15   the reverse effect where if you have a few viable

 

 16   cells with a lot of dead cells, you can actually

 

 17   get tumors developing from one cell where normally,

 

 18   you might need a million or 10 million.

 

 19             I am just wondering if you have done any

 

 20   mixtures of dead and live cells to really see how

 

 21   much is viability, how much is surrounding stuff.

 

 22             DR. MENASCHE:  No, we have not done that

 

 23   intentionally.  We have just completed a study in

 

 24   which we have looked more carefully at the patterns

 

 25   not only of cell   death, but also of cell

 

                                                               128

 

  1   proliferation.

 

  2             So, we know that we have this mix of dead

 

  3   cells and living cells, but we have not done an

 

  4   intentional mixing of them to see whether there was

 

  5   any tumor formation.

 

  6             Regarding oncogenicity, we have learned a

 

  7   lot from our colleagues working in the field of

 

  8   dystrophic myopathies, and it really seems that

 

  9   these cells have a very low tumor-retaining

 

 10   potential.

 

 11             In the newt mice in which we have injected

 

 12   our human myoblasts, we have never seen any tumor

 

 13   in spite of the fact that many of these cells

 

 14   expectedly died.

 

 15             DR. RAO:  Thank you, Doctor.

 

 16             We will move on to our next speaker, Dr.

 

 17   Epstein.

 

 18            Bone Marrow Cell Therapy for Angiogenesis:

 

 19                        Present and Future

 

 20             DR. EPSTEIN:  It is really an honor to

 

 21   have been asked to speak to this very august group.

 

 22             I wanted to emphasize because I really do

 

 23   think it is important in this, an ever-growing

 

 24   field to make sure that disclosure is presented,

 

 25   and I have a number of potential conflicts of

 

                                                               129

 

  1   interest, which I hope in no way influences what I

 

  2   will be talking about to you for the next 20 or 30

 

  3   minutes.

 

  4             I will talking about bone marrow cells and

 

  5   angiogenesis.  I wanted to start out and make a

 

  6   careful distinction.  Dr. Schneider brought this up

 

  7   in his earlier questions, but basically, what we

 

  8   are considering today is really the use of bone

 

  9   marrow cells, stem cells, progenitor cells for

 

 10   myogenesis, but also for angiogenesis.

 

 11             It is critically important to understand

 

 12   that these are very distinct targets with

 

 13   undoubtedly different mechanisms and certainly

 

 14   very, very different issues, and therefore will

 

 15   have a profound impact on how the FDA I think

 

 16   judges whether or not a particular proposal is

 

 17   meritorious.

 

 18             For example--and you have heard this very

 

 19   eloquently discussed by Dr. Menasch--for

 

 20   myogenesis, the transdifferentiation of adult

 

 21   progenitor cells or skeletal myoblasts is a

 

 22   critically important issue.  Maybe you don't need

 

 23   transdifferentiation into cardiac myocytes to

 

 24   improve myocardial contractility, but nonetheless,

 

 25   it is a very important issue to consider.

 

                                                               130

 

  1             Also, if you think about how many cells

 

  2   are present in a large myocardial scar, the issue

 

  3   of adequate numbers of cells to replace the scar to

 

  4   cause a significant biologic effect has to be

 

  5   considered, and you have heard a very eloquent

 

  6   presentation and some demonstration relating to

 

  7   this.

 

  8             Now, the issues relating to angiogenesis

 

  9   are, as I indicated a moment ago, different.

 

 10   Transdifferentiation is really not an important

 

 11   factor, because what has been recognized most

 

 12   recently is that cytocrine secretion, exerting a

 

 13   paracrine effect can induce proliferation and

 

 14   remodeling of existing vessels.

 

 15             So, it is not necessary, although it may

 

 16   happen, it is not necessary for the cells that you

 

 17   are injecting to turn into blood vessels.  They

 

 18   could induce the development of already existing

 

 19   blood vessels, and the adequate number of cells

 

 20   relating to angiogenesis is not nearly of similar

 

 21   concern secondary to these paracrine effects that

 

 22   have an amplifying activity of the individual cells

 

 23   that are injected.

 

 24             So, these are very different issues, each

 

 25   are very important.  I think the path to myogenesis

 

                                                               131

 

  1   is going to be a longer one.  I think that there

 

  2   are a lot of problems that still have to be solved,

 

  3   and I think the issue of angiogenesis, we have gone

 

  4   along that path for probably a longer period of

 

  5   time, and my sense is that we are closer to pivotal

 

  6   clinical trials even when one considers cell

 

  7   therapy, but I will be focusing my remarks on

 

  8   angiogenesis.

 

  9             The first thing I wanted to point out,

 

 10   which is obvious to anyone who is involved in the

 

 11   field now, is how complex the molecular and

 

 12   cellular mechanisms are that are involved in

 

 13   collateral formation.

 

 14             This is a slide I always like to show.

 

 15   This is a cartoon showing different genes

 

 16   expressed, either increased expression or decreased

 

 17   expression, four different times, different

 

 18   amounts, two actually wind up with a collateral.

 

 19             So, there are multiple, multiple genes

 

 20   that are necessary to actually form a new

 

 21   collateral vessel.  Just to illustrate the

 

 22   importance of interactions between different

 

 23   angiogenic cytokines, I wanted to show you the

 

 24   results of a study that we did a couple of years

 

 25   ago using a rabbit ear.

 

                                                               132

 

  1             So, here is the ear.  It is supplied by

 

  2   three major vessels.  This is a laser doppler image

 

  3   which is color coded for velocity.  Red is highest

 

  4   velocity, green is intermediate, and blue is low

 

  5   velocity.  If you tie off two of these three

 

  6   vessels, you have a marked decrease in flow, and

 

  7   the nice thing about the rabbit ear is that you

 

  8   could observe this hourly if you wanted, and you

 

  9   could with this laser doppler do repeated analyses

 

 10   of the changing flow with time.

 

 11             What you can also do is focus on a

 

 12   particular area of interest and measure tissue

 

 13   perfusion and the change in tissue perfusion that

 

 14   occurs with time.

 

 15             Here is the tying off of these vessels,

 

 16   resulting in a profound decrease in flow, which

 

 17   gradually recovers over several weeks, and in this

 

 18   particular model, it is quite interesting.  It

 

 19   plateaus off below normal flow, so this is a model

 

 20   of chronic hypoperfusion, which makes it kind of

 

 21   interesting.

 

 22             In this model, we looked at what happens

 

 23   with endogenous VEGF levels, and VEGF is a key

 

 24   angiogenic molecule, so we measured VEGF by western

 

 25   blot before the induction of ischemia, and there is

 

                                                               133

 

  1   essentially no VEGF present, however, if we measure

 

  2   VEGF levels throughout the course of this, and even

 

  3   at the end, there is a low level of VEGF present.

 

  4   So, this is further indication that we are dealing

 

  5   with a chronically ischemic preparation that has a

 

  6   background of VEGF present.

 

  7             The next issue that we wanted to document,

 

  8   we took the model during the period of chronic

 

  9   ischemia, and we take that now as our starting

 

 10   point for this experiment, where we had an

 

 11   angiopoietin-1 gene within an adenoviral vector, so

 

 12   that is the transgene, which we inject

 

 13   intradermally in the region of hypoperfusion in the

 

 14   ear.

 

 15             We inject it and we see over the course of

 

 16   time, a major increase in collateral flow and

 

 17   tissue perfusion, but remember there is background

 

 18   VEGF present. If we coinject with the adenovirus

 

 19   expressing angiopoietin-1, an inhibitor of VEGF,

 

 20   and this is a soluble VEGF receptor, so it sops up

 

 21   and inactivates whatever VEGF is present, it

 

 22   obliterates the collateral-forming effects of

 

 23   angiopoietin, so it just is an example of how you

 

 24   need multiple factors to develop your collaterals.

 

 25             If any one of these is perturbed, you

 

                                                               134

 

  1   could seriously influence the course of collateral

 

  2   development.

 

  3              Now, just as background for the cell

 

  4   therapy, there have been a number of adequately

 

  5   powered, randomized studies that have been

 

  6   performed using individual cytokines for

 

  7   angiogenesis, and basically, these are either basic

 

  8   FGF or VEGF used in the coronary circulation or the

 

  9   peripheral vasculature, either the protein was

 

 10   injected or a gene encoding the protein were

 

 11   injected.

 

 12             As of the moment, there have been no

 

 13   definitive and robust beneficial results.  There is

 

 14   trends, there is some encouragement, there is some

 

 15   early positive results, but nothing to really get

 

 16   excited about.

 

 17             Of course, as I indicated, all of these

 

 18   randomized studies to date have involved a single

 

 19   agent to promote collateral development, and it was

 

 20   these considerations about four or five years ago

 

 21   that provided the impetus for developing and

 

 22   testing a second generation of angiogenesis

 

 23   strategies, which is the use of cell therapy, which

 

 24   had the potential to deliver multiple

 

 25   collaterogenic cytokines.

 

                                                               135

 

  1             I just wanted to show this slide.  I am

 

  2   not an expert at all in stem cells, progenitor

 

  3   cells, but I just wanted to indicate to you what

 

  4   has been used in clinical trials or in late-stage

 

  5   preclinical trials.  So, hematopoietic stem cells

 

  6   characterized by positive CD34-133, which do

 

  7   progress to endothelial progenitor cells and then

 

  8   to endothelial cells, which have been shown to lead

 

  9   to an increase in collateral flow.

 

 10             Now, more recently, monocyte lineage cells

 

 11   have been demonstrated.  These are characterized by

 

 12   the lack of CD34, but having CD14 and 45 MAC-1,

 

 13   these monocyte lineage cells have been shown, not

 

 14   to produce endothelial cells directly, but

 

 15   nonetheless, are capable of inducing collateral

 

 16   formation.

 

 17             We have used freshly aspirated bone marrow

 

 18   cells  that have been filtered and directly

 

 19   injected.  These are autologous into pig ischemic

 

 20   hearts, as well as patients. You have heard about

 

 21   monocyte-derived bone marrow cells.  Dr. Perin used

 

 22   these in his study.

 

 23             We have been also working now with

 

 24   mesenchymal stem cells or stromal cells.  There are

 

 25   multiple terms that have been used to describe

 

                                                               136

 

  1   these.  These are CD34-negative, 45-negative cells,

 

  2   and these have been shown to produce collaterals.

 

  3             Now, I won't get into this in any detail,

 

  4   but you should be aware of the fact that some of

 

  5   these cells are believed to incorporate into

 

  6   developing collaterals, with that being a major

 

  7   mechanism by which they enhance the development of

 

  8   collaterals, whereas, other interventions are not

 

  9   believed to have that as a major mechanism, but the

 

 10   major mechanism being the secretion of all sorts of

 

 11   cytokines and growth factors that lead through a

 

 12   paracrine effect to the development of either new

 

 13   collaterals or the enhancement of existing

 

 14   collaterals.

 

 15              What I will be talking about, because all

 

 16   of our recent work has been done with these

 

 17   mesenchymal stem or stromal cells, I will be

 

 18   talking about that for the next few minutes, and

 

 19   these we refer to MSCs.

 

 20             So, these MSCs, just to start or justify

 

 21   our further studies, were cultured in vitro and

 

 22   assayed.  The conditioned medium was assayed, and

 

 23   here is our control cells which produce small

 

 24   amounts of VEGF MCP-1 and FGF, but the MSCs produce

 

 25   really quite large amounts of these angiogenic

 

                                                               137

 

  1   cytokines.

 

  2             So, we were very excited about that,

 

  3   thinking that  they could be little factories that

 

  4   might enhance collateral development.  So, this is

 

  5   the mouse hind limb model.  This is laser doppler

 

  6   imaging, as I showed you with the rabbit ear.  Here

 

  7   is the mouse's tail and the two hind limbs, and the

 

  8   femoral artery is ligated at day zero, and this is

 

  9   followed now every few days, and you can see there

 

 10   is some return of function under control

 

 11   conditions.

 

 12             This is just injecting media that had not

 

 13   been exposed to cells, and here is what we see with

 

 14   media alone and with the control cell.  This is

 

 15   mature aortic endothelial cells.

 

 16             But then when we inject into the hind limb

 

 17   MSCs, we see a quite marked improvement in

 

 18   perfusion, and this can be quantitated as shown in

 

 19   this slide.  So, this was a very exciting

 

 20   demonstration for us, which was repeated multiple

 

 21   times in different experiments.

 

 22             Now, I won't belabor the number of studies

 

 23   that have been done here.  It is in your handout

 

 24   that was distributed, I think it is page 20 and 22,

 

 25   but I will just go over a couple of the highlights.

 

                                                               138

 

  1             There have been 7 or probably 8 published

 

  2   studies in either chronic ischemia--and this is

 

  3   angiogenesis studies, no myogenesis--in chronic

 

  4   ischemia or in acute myocardial infarction.

 

  5             The points to be made are, number one, all

 

  6   of these studies have shown safety, they have shown

 

  7   feasibility, and they have all showed positive

 

  8   trends, they have been encouraging, but as Dr.

 

  9   Menasch very carefully pointed out with his own

 

 10   myogenesis studies, when you are dealing with such

 

 11   small numbers of patients, none of these studies

 

 12   was randomized, double-blinded.  There is no way

 

 13   you could draw any conclusions regarding efficacy.

 

 14             So, it is encouraging and it certainly

 

 15   would indicate that additional studies are

 

 16   necessary, but we can't make any inferences whether

 

 17   the strategies that work in an animal model very

 

 18   reproducibly necessarily work in humans.

 

 19             Now, I want to point out, and I think we

 

 20   have to be aware of this upfront, and any

 

 21   investigator who is involved in the field has to be

 

 22   aware of it, that there are potential problems with

 

 23   any angiogenic strategy including cell-based

 

 24   strategy.

 

 25             For example, genetics.  Here are some

 

                                                               139

 

  1   beautiful studies done by Birgit Kantor in

 

  2   collaboration with us. This is microscopic CT

 

  3   imaging of two different strains of mice. This is

 

  4   the femur, tibia, and this is the femoral artery,

 

  5   and the femoral artery had been ligated, and you

 

  6   can see that the C57 black 6 mouse has an

 

  7   extraordinary capacity to develop collaterals,

 

  8   however, about C. mouse, same ligation site, has a

 

  9   paucity of collaterals.  Well, clearly,  the same

 

 10   thing must relate to humans.

 

 11             Another thing that was raised earlier is

 

 12   the enormous variability amongst patients to

 

 13   respond to angiogenic interventions.  These are not

 

 14   patients, these are mice, and this is the typical

 

 15   experiment that I showed earlier, looking at laser

 

 16   doppler perfusion.

 

 17             The mouse has the femoral artery ligated,

 

 18   and there is a gradual recovery of flow in young

 

 19   mice, however, it you look at knockout mice that

 

 20   have high cholesterol levels, their capacity to

 

 21   develop collaterals is significantly impaired.

 

 22             Now, if you take an old mouse--these are

 

 23   mice about 18 to 20 months of age--they are really

 

 24   having trouble developing collaterals, and then if

 

 25   you take an old mouse who has high cholesterol

 

                                                               140

 

  1   levels, they are really in bad shape.

 

  2             Now, no one has demonstrated this

 

  3   relationship in humans, but I am certain that it

 

  4   occurs.  So, there is going to be different

 

  5   capacities of different individuals to develop

 

  6   collaterals, and undoubtedly reflecting different

 

  7   potential to respond to angiogenic interventions.

 

  8             Now, here is something I would like Dr.

 

  9   Schneider to look at, because he said this has not

 

 10   been published before, but it has been published.

 

 11   This is in I think JACC in 2002, but when we did

 

 12   our clinical study, injecting autologous filtered,

 

 13   freshly aspirated bone marrow cells into ischemic

 

 14   myocardium of patients, we took an aliquot of these

 

 15   cells and cultured them, and looked at VEGF

 

 16   production, as well as other cytokine production,

 

 17   and over the course of time, one sees an increase

 

 18   in VEGF production, so these cells do have the

 

 19   capacity to produce different angiogenic cytokines

 

 20   including VEGF, but that is the mean data.

 

 21             If you look at the individual data, there

 

 22   is marked patient-to-patient variability in the

 

 23   capacity to express VEGF, so here is a patient who

 

 24   really has a great capacity to produce VEGF,

 

 25   whereas, this is a patient who hardly can produce

 

                                                               141

 

  1   VEGF at all, and it is not a great stretch to think

 

  2   that this patient may not respond as vigorously to

 

  3   cell therapy as the patient whose cells have a

 

  4   great capacity to produce VEGF.

 

  5             Now, we didn't look at enough patients to

 

  6   be able to make such correlations, but I am sure

 

  7   that this is an issue that has to be addressed, as

 

  8   Dr. Schneider really pointed out before.

 

  9             Now, let's look at the cells we are

 

 10   injecting, and this was also raised earlier, so we

 

 11   are looking here at a HIF-1--I will just get into

 

 12   that in a moment--but it is a transcription factor

 

 13   that is a key modulator of the cells response to

 

 14   ischemia, so let's take it for the moment that it

 

 15   is a key angiogenic factor, so this is a Western

 

 16   showing HIF levels, and the first thing I want you

 

 17   to concentrate on is under normoxic conditions,

 

 18   young and old, HIF is not present, it is mostly

 

 19   absent as a matter of fact, in the absence of

 

 20   hypoxia.

 

 21             Now, in the young mice, if you expose

 

 22   these cells to hypoxia over 12 hours, there is a

 

 23   major increase in HIF protein, and then that has

 

 24   important compensatory effects on the cells'

 

 25   response to hypoxia, however, cells derived from

 

                                                               142

 

  1   old mice have a markedly impaired ability to form

 

  2   HIF in response to hypoxia, so there are

 

  3   age-related changes in the capacity of cells--these

 

  4   are MSCs--to perform in a way that we would expect

 

  5   them to if they were going to have a potent effect

 

  6   on collaterals.

 

  7             So, HIF is a master switch gene in the

 

  8   presence of hypoxia, a heterodimer is formed,

 

  9   HIF-1-alpha, and HIF-1-beta, which attaches to the

 

 10   promotor of many genes and turns these genes on,

 

 11   and amongst the genes are multiple genes related to

 

 12   angiogenesis, VEGF, VEGF receptor, FGF, et cetera.

 

 13             Now, just to show the biologic effects of

 

 14   what I just showed you before, that is, the

 

 15   inability to increase HIF protein in response to

 

 16   hypoxia, here are young and old mice, and we are

 

 17   looking at VEGF levels.  These are cells, MSCs

 

 18   growing in culture, and here is the intrinsic VEGF

 

 19   production.

 

 20             When we expose young cells to hypoxia,

 

 21   there is a major increase in VEGF production,

 

 22   mediated mainly by HIF-1 reduction, but old mice

 

 23   not only have the lower levels of HIF-1, but they

 

 24   have a lower target production of HIF-1, that is,

 

 25   VEGF production.

 

                                                               143

 

  1             So, these are real phenomenon I think that

 

  2   we have to be aware of and begin to start thinking

 

  3   about when we are dealing with any angiogenic

 

  4   intervention, but certainly with the cell

 

  5   therapies.

 

  6             So, these considerations provided the

 

  7   impetus to test another generation of angiogenic

 

  8   strategies, and this relates again to one of the

 

  9   earlier questions of the panelists, and we are very

 

 10   much involved in genetic manipulation of these MSCs

 

 11   to see if we could further enhance their ability to

 

 12   secrete angiogenic cytokines and so to improve

 

 13   collateral flow.

 

 14             This is a construct of Genzyme.  They have

 

 15   been very helpful in working with us in this.  In

 

 16   the absence of severe hypoxia, these two dimers of

 

 17   HIF, HIF-1-beta and alpha, there is no heterodimer

 

 18   formed because HIF-1-alpha is rapidly degraded.

 

 19             So, to overexpress HIF-1-alpha, so that

 

 20   the heterodimer can form, we transfect these cells

 

 21   with an adenoviral vector that has the HIF-1-alpha

 

 22   transgene and that has a deletion insertion,

 

 23   putting on a herpes sequence VP16, which stabilizes

 

 24   the protein under normoxic conditions, so we are

 

 25   able to overexpress HIF-1-alpha, the heterodimer

 

                                                               144

 

  1   can be formed, and the genes can be transactivated.

 

  2             So, we then looked at the capacity of this

 

  3   intervention to cause these MSCs to secrete

 

  4   angiogenic cytokines, so this is VEGF.  The cells

 

  5   now are exposed to just hypoxia, and you can see

 

  6   there is about a doubling of the amount of VEGF

 

  7   present, but when we transfect these cells with the

 

  8   HIF-1-alpha, there is a huge increase in VEGF

 

  9   production, and the same thing is true for

 

 10   fibroblast growth factor.

 

 11             So, this was really exciting to us because

 

 12   we saw that we could genetically manipulate these

 

 13   cells to make them at least in vitro more like a

 

 14   better collateral enhancer.

 

 15             We then went to our mouse ischemic hind

 

 16   limb model to test this concept.  Here are our

 

 17   control cells, mature aortic endothelial cells.

 

 18   Here are our non-transfected MSCs, and here are our

 

 19   transfected MSCs.  So, not only do we see an effect

 

 20   in vitro, but we see what would have been predicted

 

 21   from the in vitro effects in vivo.

 

 22             I think that this is probably something

 

 23   that we have to think about given the effects of

 

 24   various risk factors on the ability of cells to

 

 25   achieve their desired effects.

 

                                                               145

 

  1             I did want to point out for the panel that

 

  2   down the line, we are not only going to be talking

 

  3   about cells, but cell-derived products in

 

  4   cardiovascular therapy, and I will just spend a

 

  5   moment on this, and mention the effects of

 

  6   conditioned medium on collateral development.

 

  7             As I showed you before, if you grow cells

 

  8   in culture and allow them to produce whatever

 

  9   goodies they are producing, and then you take the

 

 10   media and you inject that media into the ischemic

 

 11   hind limb of mice, you get--well, that will be the

 

 12   next slide--but here is what I showed you before,

 

 13   so these in the media contains more VEGF, more

 

 14   MCP-1, more FGF, and multiple other gene products

 

 15   that we haven't tested, but when we put this

 

 16   conditioned medium into the ischemic hind limb of

 

 17   the mouse, here is the control again, here is our

 

 18   control.

 

 19             Here is the injection of the MSC

 

 20   conditioned medium.  We see that the media alone

 

 21   has the capacity to increase collaterals.  So, this

 

 22   undoubtedly is going to be something that you are

 

 23   not going to see an application to this, I don't

 

 24   think in the next few months, but in the next six

 

 25   months or a year, I think that the cell products is

 

                                                               146

 

  1   another very interesting way to use the angiogenic

 

  2   potential that bone marrow cells have.

 

  3             Just to show that this is biologically

 

  4   important, we looked at the number of collateral

 

  5   vessels the media increased, the number of

 

  6   collateral vessels, the strength of the leg as an

 

  7   ambulatory score, the media increased that, and

 

  8   also the amount of atrophy that occurs in the calf

 

  9   as a result of ischemia, and the media injection

 

 10   decreases that, so it was a biologically relevant

 

 11   intervention.

 

 12             I just wanted to mention safety concerns,

 

 13   and I was interested in Dr. Menasch's comment

 

 14   about this.  There are multiple well-known ones

 

 15   that are usually tracked, and I won't get into

 

 16   this, but I would just alert you to something that

 

 17   is more theoretical than proven, but I think you

 

 18   have to be aware of it and at least consider it

 

 19   when you are considering the safety of angiogenic

 

 20   interventions.

 

 21             That is--and this is a general rule that I

 

 22   have come up with--whatever induces angiogenesis,

 

 23   induces atherogenesis, and I refer to this as the

 

 24   Janus phenomenon. Janus was a Greek god with two

 

 25   heads, so that when he looks out one way, he is

 

                                                               147

 

  1   also looking at the other.  That goodness can also

 

  2   be badness, there are no free lunches.

 

  3             So, I would think that any angiogenesis

 

  4   intervention, one of the potential side effects

 

  5   that one should look for is the acceleration of the

 

  6   atherogenic process.

 

  7             This is just a slide that is still in

 

  8   development, but basically, it shows that when you

 

  9   induce ischemia in a hind limb, you have decreased

 

 10   PO2.  This activates cytokine release, which

 

 11   activates bone marrow cells, splenocytes, many

 

 12   inflammatory cells.

 

 13             We now know that inflammatory cells are

 

 14   critically important to the development of

 

 15   collaterals.  Macrophages have been shown to be

 

 16   critical.  We have shown that both CD4,

 

 17   t-lymphocytes, and CD8 t-lymphocytes are critical

 

 18   to collateral development, but these same factors,

 

 19   these same inflammatory factors also have been well

 

 20   described to lead to, much longer than

 

 21   angiogenesis, an acceleration of atherosclerosis.

 

 22             So, if it causes angiogenesis, think very

 

 23   hard as to whether it might be worsening the

 

 24   atherosclerotic process.

 

 25             The conclusions:  single molecule-based

 

                                                               148

 

  1   strategies to improve collateral flow, although

 

  2   effective in animals, have yet to be proven

 

  3   efficacious in patients.  Cell-based strategies

 

  4   have great promise because of the ability of bone

 

  5   marrow derived progenitor cells to secrete multiple

 

  6   collaterogenic cytokines.

 

  7             However, cell-based therapies also have

 

  8   the potential of achieving suboptimal effects

 

  9   because of the effects of aging and other risk

 

 10   factors on cell function.  The optimal strategy has

 

 11   yet to be identified, but genetic manipulation of

 

 12   cells would appear to hold great promise, and use

 

 13   of cell products, such as conditioned medium

 

 14   derived from cells, will also undoubtedly be

 

 15   explored as a therapeutic strategy in the near

 

 16   future.

 

 17             Thank you.

 

 18             [Applause.]

 

 19             DR. RAO:  Thank you, Doctor.

 

 20             We have time for a few quick questions.

 

 21                               Q&A

 

 22             DR. SCHNEIDER:  Steve, as an exploratory

 

 23   tool, conditioned medium for angiogenesis makes a

 

 24   lot of sense for the reasons that you articulated,

 

 25   but as a therapeutic product, that would be true

 

                                                               149

 

  1   if, and only if, conditioned medium contained

 

  2   products that could not be identified or could not

 

  3   be added combinatorily from defined factors.

 

  4             So, it seems to me it will be especially

 

  5   useful in those conditioned medium experiments to

 

  6   test the effect of specific blockers and find out,

 

  7   at a reductionist level, what the components are.

 

  8   If it were as simple as angiopoietin and VEGF, one

 

  9   could use angiopoietin and VEGF.

 

 10             DR. EPSTEIN:  Right, it's a very good

 

 11   point.  My own feeling is, having been in this

 

 12   field now for 12--more than that--14 years, it is

 

 13   so complex and the number of factors that are

 

 14   involved in collateral development are not 4 or 5,

 

 15   but they are dozens, and maybe even more than that,

 

 16   that I personally will not waste time trying to

 

 17   figure out what two products are enough or what

 

 18   three products, I don't believe that, but the

 

 19   cells, they know how to develop collaterals, I mean

 

 20   they are doing it all the time, so I will go with

 

 21   cell therapy, and I will allow someone else to look

 

 22   at what combination of three factors might be

 

 23   optimal.

 

 24             It may be that you could find such

 

 25   factors, but don't forget, not only do you have to

 

                                                               150

 

  1   know what factors are present, but you have to know

 

  2   in what concentrations, and so on.

 

  3             I think the cells, they are eliminating so

 

  4   many of the issues that if we were going to look at

 

  5   individual cytokines, we would have to explore for

 

  6   years, so it is a good point, but I think that the

 

  7   practical issues, given the huge complexity of

 

  8   this, would be overwhelming.

 

  9             DR. SCHNEIDER:  To follow up on your

 

 10   comment, which I would share, that it is extremely

 

 11   likely that engineered cells will outperform naive

 

 12   cells,  I would like to ask the participants from

 

 13   FDA what additional hurdles are seen in the

 

 14   consideration of gene-engineered cells to be

 

 15   applied to these therapeutic situations.

 

 16             DR. NOGUCHI:  I think we can answer in

 

 17   general that actually, we have a fairly rich

 

 18   experience with gene-modified cells that have been

 

 19   given to individuals for a whole variety of

 

 20   diseases, not too many for cardiovascular, but I

 

 21   wouldn't expect that we would have very much

 

 22   difficulty in really being able to handle that.

 

 23             DR. RAO:  Dr. Mul.

 

 24             DR. MULE:  Combining your presentation

 

 25   with Dr. Menasch's, I was sitting here wondering

 

                                                               151

 

  1   what is known about, if one takes whole bone marrow

 

  2   cells and perhaps Dr. Kurtzberg can add to this,

 

  3   with Dr. Menasch's studies, if there are cells

 

  4   within the marrow that can give rise to myotubes,

 

  5   and you combine that with a population of cells

 

  6   that could be responsible for collaterogenesis, the

 

  7   issue is are they the same cell or are we at a

 

  8   period in time where  we can identify two subsets

 

  9   within the marrow that conceivably could be

 

 10   combined to overcome some of the issues of

 

 11   viability that Dr. Menasch has talked about.

 

 12             If the answer is no, we are not there yet,

 

 13   then, it begs the question if one were to use an

 

 14   adenovirus to introduce a gene to improve

 

 15   collaterogenesis into the cell population, that is

 

 16   identifiable for producing myotubes, the issue is

 

 17   does that manipulation adversely impact the ability

 

 18   of that cell to create myotubes.

 

 19             DR. EPSTEIN:  Well, those are sensational

 

 20   questions, and I have never thought of this last

 

 21   one before, but it is certainly--you know, it is so

 

 22   easy to do harm, and it is so hard to do good, so I

 

 23   mean your question is very apt, I mean does the

 

 24   very expression of the cytokines that enhance

 

 25   angiogenesis, might it interfere with myogenic

 

                                                               152

 

  1   potential, I don't think anybody has done that

 

  2   experiment. Hopefully, the answer will be no, but

 

  3   it certainly is an experiment that has to be done.

 

  4             The MSCs that we are deriving from the

 

  5   bone marrow do not differentiate into myoblasts,

 

  6   and it would be a very interesting experiment to

 

  7   take Dr. Menasch's approach and mix these in with

 

  8   the skeletal myoblasts to see, because I can't

 

  9   understand how, if you have a scar, and you are

 

 10   injecting cells into the scar, and you do nothing

 

 11   about the blood flow, why those cells won't turn

 

 12   into scar.  The blood supply clearly was

 

 13   demonstrated to be inadequate because you have got

 

 14   scar.

 

 15             Of course, Dr. Menasch is actually doing

 

 16   some experiments now using the same molecule that

 

 17   we are using to induce collateral formation, so it

 

 18   certainly is a very, very important strategy to

 

 19   test.

 

 20             DR. KURTZBERG:  In answer to the other

 

 21   question raised, I don't personally think we know

 

 22   which subsets are important yet, or whether subsets

 

 23   are more important than whole cell preparations.  I

 

 24   think that all should be the focus of questions

 

 25   going forward.

 

                                                               153

 

  1             Evan Snyder has an interesting model of

 

  2   spinal cord injury and repair.  It's a rat model.

 

  3   They ligate, take a hunk of spinal cord and then

 

  4   inject allogeneic cells and look at repair, and

 

  5   they see repair and re-formation and re-connection

 

  6   of nerves, but when they went back and looked to

 

  7   see what cells did it, it turned out they were host

 

  8   cells that were facilitated by something that the

 

  9   allogeneic cells brought to the table, although

 

 10   they don't know what.

 

 11             To me, that just points out how much we

 

 12   don't know and how complex the process is, and how

 

 13   much more we need to study.

 

 14             DR. RAO:  One last question.  To me, and I

 

 15   am somewhat naive in this field, there is a

 

 16   difference between new vessel initiation and

 

 17   collateral formation of regrowth in terms of the

 

 18   factors which had acquired, and so on, and it seems

 

 19   to me in some models of cardiac ischemia, what we

 

 20   are looking at are completely ischemic regions and

 

 21   long term, which there is no regrowth, and if you

 

 22   had to do anything, it would be new vessel

 

 23   formation.

 

 24             Would it be fair to say that we can't

 

 25   extrapolate from the current models that you talked

 

                                                               154

 

  1   about in terms of the religation and

 

  2   revascularization, or is it reasonable to be able

 

  3   to extrapolate from those models to what you think

 

  4   might happen in a cardiac model?

 

  5             DR. EPSTEIN:  Well, I think it is

 

  6   reasonable to extrapolate because we demonstrate

 

  7   that we are able to improve perfusion, but your

 

  8   question is really a very interesting one, and that

 

  9   is, there used to be a major emphasis that

 

 10   increased perfusion is just due to angiogenesis or

 

 11   the development of new capillaries.

 

 12             Well, I think most people involved in the

 

 13   field would agree at this point that capillaries

 

 14   don't increase flow.  They facilitate the

 

 15   distribution of flow, and what you need is an

 

 16   increase in conductance vessels or arteriogenesis

 

 17   to truly produce an overall increase in flow.

 

 18             However, we have some preliminary data to

 

 19   suggest that both processes are real, that

 

 20   angiogenesis is a part of arteriogenesis, and that

 

 21   you need the development of capillaries, that the

 

 22   development of new vessels, capillaries, can

 

 23   remodel to form collaterals.

 

 24             That is why I don't use the term anymore

 

 25   of angiogenesis.  I say "collaterogenesis," because

 

                                                               155

 

  1   it gets away from the mechanistic aspects, which

 

  2   are critically important, and we still don't have

 

  3   the answer what cytokines produce angiogenesis,

 

  4   what are important in terms of arteriogenesis, and

 

  5   are both important to actually optimize the

 

  6   development of collaterals, so we still have a

 

  7   couple of years I think to go to answer that

 

  8   question.

 

  9             DR. RAO:  One last question

 

 10             DR. HARLAN:  When you showed the

 

 11   adenoviral HIF transfected cell lines and showed

 

 12   that those were more efficient at elaborating

 

 13   cytokines and inducing vessel growth, it adds

 

 14   another question, that then becomes, however,

 

 15   potentially anyway, a less well refined product,

 

 16   the cell-conditioned medium from those cells, and

 

 17   in view again of what we heard when we started

 

 18   today, as we move forward with thinking about

 

 19   delivering products to people, you want them to be

 

 20   defined, and I wonder if you would comment on that.

 

 21             DR. EPSTEIN:  Well, if this field is to

 

 22   move forward, I think that criteria is going to

 

 23   have to be eliminated because there is no way you

 

 24   are going to be able to define--maybe I am

 

 25   exaggerating--the hundreds of molecules that these

 

                                                               156

 

  1   cells are producing.  We don't even know how to

 

  2   measure them.

 

  3             But I am sure the FDA allows the infusion

 

  4   of serum and plasma from one individual to another.

 

  5   Do you know what is in that serum?

 

  6             DR. HARLAN:  I am not the FDA.

 

  7             [Laughter.]

 

  8             DR. EPSTEIN:  So, there is a precedent for

 

  9   not knowing what you are injecting.  To be honest,

 

 10   we are injecting cells, and we know a few of the

 

 11   molecules that they are secreting, but we have no

 

 12   idea of the concentration, and whether it is going

 

 13   to vary from one patient to another, and if a

 

 14   patient has diabetes or has hypercholesterol, so

 

 15   believe me, if you going to be compulsive and say

 

 16   we have to know the concentration, when, over the

 

 17   course of time, those molecules are up, and what is

 

 18   their interaction, we have to stop this field right

 

 19   now, it can't move forward, and it is too bad.  I

 

 20   mean you would like to know everything, but this is

 

 21   not a characteristic of cell therapy.

 

 22             DR. RAO:  On that note, we will break for

 

 23   lunch.  We broke a little late, so we will try and

 

 24   come back a little bit later, but not too much, so

 

 25   we will shoot for 1:00.

 

                                                               157

 

  1             [Whereupon, at 12:15 p.m., the proceedings

 

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

 

                                                               158

 

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

 

  2                                                    [1:12 p.m.]

 

  3             DR. RAO:  Good afternoon.

 

  4             Before we begin with the talks, I would

 

  5   like to introduce three more members of the

 

  6   committee who have just joined us.  I like to let

 

  7   them do it.

 

  8             DR. HIGH:  My name is Katherine High.  I

 

  9   am on the faculty at the University of

 

 10   Pennsylvania.  I am a hematologist with an interest

 

 11   in gene transfer for hematological disease.

 

 12             DR. BLAZER:  My name is Bruce Blazer.  I

 

 13   am at the University of Minnesota in the Department

 

 14   of Bone Marrow Transplantation with an interest in

 

 15   immunobiology.

 

 16             DR. RAO:  We also have Dr. Grant from the

 

 17   FDA.

 

 18             DR. GRANT:  Hi.  I am Steve Grant.  I am a

 

 19   cardiologist.  I am also a clinical reviewer within

 

 20   the Office of Cellular Tissue and Gene Therapies.

 

 21             DR. RAO:  We will continue with the series

 

 22   of talks that were scheduled.

 

 23             The next speaker is going to be Dr.

 

 24   McFarland.

 

 25              Cellular Therapies for Cardiac Disease

 

                                                               159

 

  1             DR. McFARLAND:  Thank you, Dr. Rao, and

 

  2   welcome back from lunch.

 

  3             [Slide.]

 

  4             This slide is intended to remind me to

 

  5   answer the implicit question which may have been

 

  6   raised, and the question is:  Isn't the FDA putting

 

  7   the cart before the horse?

 

  8             The answer is, well, yes, in a way.  We

 

  9   thought that it would be good to focus and give

 

 10   people a peek at what is in the cart below those

 

 11   flat-screen monitors, I suppose, before we spend

 

 12   the afternoon dealing with the horse which is

 

 13   pulling the cart, the draft horse of product

 

 14   development being preclinical studies and product

 

 15   characterization.

 

 16             [Slide.]

 

 17             As Dr. Rao said, I am Richard McFarland,

 

 18   and I am in the Office of Cell Tissue and Gene

 

 19   Therapy in CBER.

 

 20             [Slide.]

 

 21             What I am going to do, I have been charged

 

 22   with providing a perspective, FDA perspective to

 

 23   the preclinical and manufacturing issues of cell

 

 24   therapies for cardiac diseases.

 

 25             First, I am going to describe the general

 

                                                               160

 

  1   framework in which the FDA conducts our

 

  2   science-based assessment of safety of novel

 

  3   cellular therapies prior to allowing clinical

 

  4   trials to proceed.

 

  5             Second, I am going to describe the goals

 

  6   of preclinical testing, safety testing in general,

 

  7   and then a little specific about how that applies

 

  8   to cellular therapies for cardiac disease, and

 

  9   finally introduce the speakers for the rest of the

 

 10   afternoon.

 

 11             [Slide.]

 

 12             FDA review is product-based, and it

 

 13   parallels prudent product development.  This is in

 

 14   contra-distinction   to the NIH grant process,

 

 15   which is more based on diseases  and organ systems,

 

 16   which is illustrated just in the administrative

 

 17   structure of the two agencies, NIH being primarily

 

 18   divided by institutes, and FDA being primarily

 

 19   divided by products that we regulate.

 

 20             That means that our FDA review is

 

 21   dependent on the characteristics of a specific

 

 22   product, and the preclinical studies are designed

 

 23   to support the use of specific products, and the

 

 24   clinical trial design that we review is designed to

 

 25   be supported by manufacturing and preclinical data.

 

                                                               161

 

  1             That product-based review is framed by

 

  2   regulations.  I think I am the one designated to

 

  3   get ready for regulations from the FDA.

 

  4             [Slide.]

 

  5             These are selections from the

 

  6   Investigational New Drug regulations.  I want to

 

  7   highlight a few things, that being that regulations

 

  8   stipulate there is adequate information about

 

  9   pharmacological and toxicological studies, that the

 

 10   sponsor has concluded that it is reasonably safe,

 

 11   and that the kind, duration, and scope of those

 

 12   required tests vary with the nature of the proposed

 

 13   clinical investigations.

 

 14             [Slide.]

 

 15             A little further down in the regs, for

 

 16   each toxicology study that is intended primarily to

 

 17   support the   safety of the proposed clinical

 

 18   investigation, a full tabulation of data suitable

 

 19   for detailed review should be submitted.

 

 20             This is kind of critical to the way that

 

 21   we do the review in that we need to get enough data

 

 22   to do a detailed review.

 

 23             [Slide.]

 

 24             Let's back up for a minute and go to

 

 25   preclinical evaluation in general.  What are the

 

                                                               162

 

  1   goals of preclinical evaluation with a perspective

 

  2   of supporting trials?

 

  3             One is to provide a rationale for the

 

  4   proposed therapy, discern mechanism of action,

 

  5   identify "at risk" patient populations, recommend

 

  6   safe starting doses and escalation schemes for

 

  7   humans, do a preliminary risk/benefit assessment,

 

  8   and to identify parameters for potential clinical

 

  9   monitoring.

 

 10             [Slide.]

 

 11             I will talk a little more specifically

 

 12   about use of preclinical models for cellular

 

 13   therapies.  Preclinical models are used to provide

 

 14   the scientific rationale with the cellular product

 

 15   intended for clinical use, to understand cell

 

 16   function, trafficking, and differentiation as all

 

 17   these factors impinge on safety, as well as

 

 18   modeling of routes of administration.

 

 19             [Slide.]

 

 20             If we had an ideal animal model for

 

 21   cardiac cell therapies, it would have a similar

 

 22   pathophysiology to humans that would improve the

 

 23   predictability of human risk from the models,

 

 24   similar anatomy to humans, which would allow us to

 

 25   use various routes of delivery including catheters

 

                                                               163

 

  1   of various types, with actually the clinical

 

  2   catheter, it would also allow us to do extensive

 

  3   dose exploration of cells, and it would be

 

  4   immune-tolerant to human cells, so you could

 

  5   actually use a human cellular product.

 

  6             [Slide.]

 

  7             Well, such an animal model doesn't really

 

  8   exist, so we often use  syngeneic animal models of

 

  9   cardiac diseases because they can provide us useful

 

 10   data for assessment of safety.

 

 11             That would be cells from analogous cell

 

 12   source processed in a similar way from animals,

 

 13   autologous cells in the animal species or syngeneic

 

 14   species, gives rise to potential processing,

 

 15   formulation, and storage differences, and limited

 

 16   product characterization both preclinically and

 

 17   clinically leads to some uncertainty in addition to

 

 18   the uncertainty that is inherent in the modes

 

 19   themselves.

 

 20             [Slide.]

 

 21             Add to that, we have added complexity due

 

 22   to innovative delivery systems, many of which have

 

 23   not been tested for delivery of cells.  Common

 

 24   delivery systems that we have seen, intraoperative

 

 25   transepicardial injection usually during CABG,

 

                                                               164

 

  1   catheter-mediated transendocardial injection, and

 

  2   catheter-mediated via cardiac vein, all of which I

 

  3   think were discussed this morning.

 

  4             [Slide.]

 

  5             I only want to present a short scaffold of

 

  6   the animal models that have been published so far,

 

  7   and our speakers this afternoon will much more

 

  8   extensively discuss the data.

 

  9             There have been data in small animal

 

 10   models, often cryoinjury, occasionally coronary

 

 11   artery ligation to give an  ischemic area damaged

 

 12   myocardium.   One of the advantages of the small

 

 13   animal systems is that you have them available, at

 

 14   least in mouse and rat, to use a human cellular

 

 15   product to give you an idea of function and safety

 

 16   of those cells. Primarily, it has been in mouse,

 

 17   rat, and rabbit.

 

 18             [Slide.]

 

 19             Large animal models, typically, dog,

 

 20   sheep, and pig, and we have seen some of those data

 

 21   this morning, as well, an ameroid constrictor used

 

 22   to generate an ischemic area has been a popular

 

 23   model of disease.  They are amenable to catheter

 

 24   administration and more amenable  to clinical

 

 25   monitoring modalities, however, you are stuck with

 

                                                               165

 

  1   using syngeneic cells in this situation.

 

  2             [Slide.]

 

  3             The regulations say that the data should

 

  4   be adequate and extensive.  What are the potential

 

  5   sources of data to support initiation of clinical

 

  6   trials?

 

  7             Well, the gold standard would really be

 

  8   preclinical studies specifically designed to

 

  9   support a specific trial with a specific cell.

 

 10             We also have data from other potential

 

 11   sources:   existing animal studies that were

 

 12   designed to answer other questions, in-vitro

 

 13   studies, clinical trials using the "same" product.

 

 14             [Slide.]

 

 15             However, we use data that are published

 

 16   and unpublished.  Using published data either from

 

 17   animal studies or human studies as sole support for

 

 18   initiation of clinical trials raises some

 

 19   questions, some complexities, because often these

 

 20   studies are not designed to answer a toxicologic

 

 21   question, and therefore, adequate toxicology

 

 22   endpoints may not have been incorporated into the

 

 23   design.

 

 24             If they were incorporated into the design,

 

 25   but not in the publication, we need access to those

 

                                                               166

 

  1   data.  Published reports may provide sufficient

 

  2   information for independent review.

 

  3             [Slide.]

 

  4             There are some limitations in using

 

  5   published studies.  Protocols in the studies need

 

  6   to be sufficiently detailed.  We need to be able to

 

  7   do our independent review as per our regulations.

 

  8   We need to see specifics of the route of

 

  9   administration.

 

 10             We need to see catheter specifics, such as

 

 11   identity of the catheter, flow rate, pressures,

 

 12   effects of catheters on cells, location of

 

 13   injection in relation to the ischemic area, and

 

 14   protocols, either animal studies or human

 

 15   protocols, we need the control details of the

 

 16   "routine" monitoring and analytical plans.

 

 17             [Slide.]

 

 18             The data must be presented in sufficient

 

 19   detail.

 

 20             In-process and lot-release data from

 

 21   manufacturing need to be presented in sufficient

 

 22   detail for us to know exactly what the product is,

 

 23   and complete study reports for both animal and

 

 24   clinical studies.

 

 25             [Slide.]

 

                                                               167

 

  1             Cellular products used in published

 

  2   reports may not be comparable to the intended

 

  3   clinical product.  Often, in published reports,

 

  4   there is insufficient data to allow us to make a

 

  5   comparability assessment, and that is either

 

  6   because the data don't exist or due to editorial

 

  7   constraints of the publication.

 

  8             [Slide.]

 

  9             So, given the limitations of the framework

 

 10   or the window at which FDA is required to look at

 

 11   these, and the detail that we are required to look

 

 12   for prudent product development raises some

 

 13   regulatory challenges.

 

 14             These are rather recurrent regulatory

 

 15   challenges.  This is does the submission contain

 

 16   sufficient information to assess risk to the

 

 17   subjects in the proposed trial.  It is a question

 

 18   that we ask at the end of our review.  Were

 

 19   adequate preclinical studies performed?  If they

 

 20   were performed, were the data submitted in

 

 21   sufficient detail to conduct an independent review?

 

 22             If sufficient data are present, then we

 

 23   get to the question, are the risk to human subjects

 

 24   reasonable and significant?

 

 25             [Slide.]

 

                                                               168

 

  1             That gives you an idea of the framework

 

  2   with which we need to look through to the window of

 

  3   science and what we are obligated to do as we make

 

  4   an assessment.

 

  5              There is discretion, there is ability to

 

  6   be flexible within the regulations, and what we are

 

  7   asking the committee to do over these two days is

 

  8   to give us an idea of what the state of the science

 

  9   is.  It will be reflected in the questions that we

 

 10   will be discussing tomorrow afternoon.

 

 11              What is the state of the science?  What

 

 12   is a reasonable amount of data for us to be looking

 

 13   at?

 

 14             This afternoon, we are going to have two

 

 15   speakers that  are focusing primarily on cells.

 

 16   Doris Taylor from the University of Minnesota and

 

 17   Silviu Itescu from Columbia.

 

 18             After the break, followed by Dr. Nick

 

 19   Jensen from the Center for Devices and Radiologic

 

 20   Health at the FDA, who will focus on delivery

 

 21   devices and some of the issues that are related to

 

 22   development and testing of delivery devices, which

 

 23   are an integral part of our preclinical

 

 24   development.

 

 25             Then, Robert Lederman from the NIH will be

 

                                                               169

 

  1   discussing some of his experiences from being in

 

  2   the trenches of doing studies with devices, cardiac

 

  3   diseases.

 

  4             DR. RAO:  Thank you.

 

  5             We will now have Dr. Taylor.

 

  6                       Guest Presentations

 

  7             Myoblasts:  The First Generation Cells for

 

  8              Cardiac Repair:  What Have We Learned

 

  9             DR. TAYLOR:  Thank you.  I have to confess

 

 10   that if I had seen those previous slides before I

 

 11   had signed my talk, it would be a completely

 

 12   different talk, so bear with me.

 

 13             I am going to be talking about some of our

 

 14   data and some of the data from the rest of the

 

 15   field, but I think what I really want to focus on

 

 16   is myoblasts for cardiovascular repair and what

 

 17   lessons we can learn from the cells that have been

 

 18   used for the longest period of time preclinically,

 

 19   and I think another way to think about this is gene

 

 20   therapy revisited, are we going to do it all over

 

 21   again.

 

 22             I think the point that I want to make is

 

 23   that there are a lot of lessons that we can learn

 

 24   from the gene therapy field as we are going forward

 

 25   with cell therapy, and I think it is important to

 

                                                               170

 

  1   take those lessons away from this.   I will be glad

 

  2   to talk about that in more detail if people have

 

  3   questions later.

 

  4             I like to start with this because this is

 

  5   what I tell the people in my lab, and I think it is

 

  6   true - make everything as simple as it is, and no

 

  7   simpler, from Einstein.

 

  8             Philippe will recognize this.  This is an

 

  9   image I borrowed from his Lancet manuscript in

 

 10   2001, showing the first patient into whom myoblasts

 

 11   were actually delivered clinically.  I actually use

 

 12   it to illustrate what I think is the salient point

 

 13   here, which is that most of us are dealing with

 

 14   animals as well as in patients, infarcted

 

 15   myocardium, where a process of events has occurred

 

 16   that starts with inflammation, moves to cardiocyte

 

 17   apoptosis, a remodeling and compensation process

 

 18   that you heard about in extreme detail this

 

 19   morning, scar expansion, decompensation, and

 

 20   progression to failure.

 

 21             The truth of the matter is we are trying

 

 22   to intervene in this with either cells, genes, or

 

 23   devices, but we don't know where in this cascade we

 

 24   are actually intervening, nor do we know where we

 

 25   should be intervening.

 

                                                               171

 

  1             I think most of us who got into this field

 

  2   envisioned it, first, as a field where we would

 

  3   intervene early after an acute myocardial

 

  4   infarction to try to prevent the slippery slope

 

  5   here of remodeling, scar expansion, decompensation,

 

  6   and failure, but the truth of the matter is that

 

  7   most of the patients in whom studies have been done

 

  8   are patients who have already progressed to some

 

  9   degree of failure.

 

 10             Although we initially started to begin to

 

 11   look at prevention and repair of not only cardiac

 

 12   injury, but also vascular injury, we are now really

 

 13   trying to understand whether or not we can move

 

 14   back up this scale in a reverse remodeling way, or

 

 15   to grow new cells, and I think those are the

 

 16   questions that are really out there in the field

 

 17   right now.

 

 18             The Holy Grail in this field then is that

 

 19   transplanted cells cannot not only engraft, but

 

 20   restore blood flow and contractility to injured

 

 21   myocardium, and all of you know, because you have

 

 22   seen some of the data from Philippe and are fairly

 

 23   well versed in this field or you wouldn't be here,

 

 24   that there is more than 15 years of preclinical

 

 25   data in rabbit and dog, there is at least 1 to 5

 

                                                               172

 

  1   years of preclinical data in pig, rat, mouse,

 

  2   sheep, and now hamster, all of which showed in our

 

  3   preclinical models that transplanting autologous

 

  4   skeletal muscle derived cells was safe, effective,

 

  5   and feasible, and therefore, Phase I, surgical and

 

  6   intervascular studies were initiated worldwide.

 

  7             I think the future that we will probably

 

  8   ultimately all try to examine is what is ultimately

 

  9   the best cell for cardiac repair.  I have been

 

 10   asked to focus on myoblasts, so I am going to do

 

 11   that although a little bit later this afternoon, I

 

 12   am going to talk necessarily a little bit about

 

 13   comparisons among cell types because I think it

 

 14   really begins to ask questions about mechanism

 

 15   that we have to address as we are going forward.

 

 16             Obviously, the best cell may be some

 

 17   autologous bone marrow-derived product.  It may be

 

 18   a cell plus or minus a therapeutic gene to either

 

 19   promote angiogenesis or some other signaling

 

 20   cascade that maybe promote cell survival.

 

 21   Ultimately, it may be embryonic stem cells although

 

 22   I would submit at least in this country, we are a

 

 23   number of years away from those cells, not only

 

 24   because we don't understand how to regulate their

 

 25   differentiation, but because we also don't

 

                                                               173

 

  1   understand how to make them stop dividing

 

  2   appropriately.

 

  3             Then, obviously, you can't be from the

 

  4   University of Minnesota without talking about

 

  5   adult-derived stem cells.

 

  6             So, what are the questions in the field?

 

  7   I am just going to put my opinions out there.  I

 

  8   think just keep these in mind as a background as we

 

  9   go forward.

 

 10             Is there a best cell?  I don't think there

 

 11   is a best cell.  I think it really depends on the

 

 12   patients, the time after injury, the dose and a

 

 13   number of other factors.

 

 14             Is there just a better way to get it

 

 15   there?  One of the questions that we keep coming

 

 16   back to over and over and over is whether or not

 

 17   cell therapy is just going to ultimately be another

 

 18   local drug delivery problem, and whether or not we

 

 19   are really going to be able to get the cells to

 

 20   where we need them or whether they have the

 

 21   capacity to actually migrate there or home there,

 

 22   and I think that is going to be an important

 

 23   phenomenon to begin to explore.

 

 24             The other side of that is do the cells go

 

 25   where we want them, or do the cells go where we

 

                                                               174

 

  1   want them and somewhere else where we don't want

 

  2   them, and I think we will come back to that.

 

  3             Should we just use growth factors and

 

  4   forget cells?  I am not going to focus on that.

 

  5             Is there a future for biologic devices,

 

  6   and is the real question dose, timing, and choosing

 

  7   the right patients for the right cell?  I would

 

  8   submit that it probably is.

 

  9             So, where are we in this field?  Well,

 

 10   this is a table that I copied from a review by

 

 11   Loren Field, and the table goes on for slides, just

 

 12   to show that in terms of preclinical myoblast

 

 13   transplantation, there is a huge amount of data out

 

 14   there, and what the data really begin to show is

 

 15   that there are a lot of different cells that have

 

 16   been used, there are a lot of different species

 

 17   that have been used from mouse, rat, rabbit, pig,

 

 18   dog, and sheep, that these cells have been injected

 

 19   either into normal heart, cryo-injured heart,

 

 20   hearts where vessels have been occluded, and that

 

 21   surprisingly, most people didn't actually measure

 

 22   improvements in function or in angiogenesis or in

 

 23   survival.

 

 24             I think that is important to consider as

 

 25   we really try to pull together the summary of data

 

                                                               175

 

  1   from myoblast cell therapy.

 

  2             So, how do we really do this?  Well, for

 

  3   myoblasts, you basically take a chunk of muscle and

 

  4   you grow cells in vitro, and you end up with cells

 

  5   in a dish, and then you deliver these cells to the

 

  6   injured myocardium and you measure the effect.

 

  7             So, you inject them, you deliver them, and

 

  8   you measure the effect.  So, I am going to go

 

  9   through the different parameters here, talking

 

 10   primarily about myoblasts and what exists in each

 

 11   of these areas.

 

 12             So, the cells are typically referred to as

 

 13   myoblasts, but the bottom line is these are

 

 14   muscle-derived cells that contain, not only

 

 15   myoblasts, but also often more fibroblasts than

 

 16   myoblasts, a number of cells called SP or site

 

 17   population cells, and then a whole lot of other

 

 18   cells that we don't necessarily know how to

 

 19   characterize yet.

 

 20             So, this is a very heterogeneous cell

 

 21   population, and you do take these cells and you

 

 22   grow them in a dish for several weeks, and what

 

 23   that means is that all of us are exposing these

 

 24   cells to serum-containing medium, and that what

 

 25   Arnold Kaplan learned years ago from mesenchymal

 

                                                               176

 

  1   cells is that when you are using serum, what is in

 

  2   that serum, FDA regulations notwithstanding,

 

  3   matters, and that the lot number of serum is going

 

  4   to give you a different outcome in terms of the

 

  5   numbers of these cells and what their phenotype is

 

  6   when you are done at the end of the day.

 

  7             In terms of injury models where myoblasts

 

  8   have been delivered, the primary injury models are

 

  9   either acute myocardial infarction, acute being

 

 10   anywhere from a week to one month, and primarily

 

 11   that has been a cryoinjury model or coronary artery

 

 12   ligation.

 

 13             The question that really arises--and then

 

 14   those have varied dramatically in size, the timing

 

 15   after creation of this infarction to the delivery

 

 16   of cells matters, and inflammation, so are these

 

 17   the same as a clinically relevant injury?  Well,

 

 18   the size differs, the timing differs, and the

 

 19   inflammation differs with regard to what is

 

 20   actually seen in patients.

 

 21             In terms of cells, the myoblasts,

 

 22   primarily how they have been delivered, more than

 

 23   90 percent of the cells preclinically and

 

 24   clinically have been delivered by surgical

 

 25   approach.

 

                                                               177

 

  1             Some preclinical data exists in terms of

 

  2   delivering cells via catheter in a pig, but again

 

  3   there are open questions about dose, about where

 

  4   those cells are delivered, about when those cells

 

  5   are delivered, and there is virtually no data out

 

  6   there about the vehicle in which the cells are

 

  7   given.

 

  8             Typically, people either say they inject

 

  9   the cells in saline or they inject them in the cell

 

 10   growth medium minus the serum.

 

 11             As I said up here, lot number matters, so

 

 12   the vehicle differs dramatically.

 

 13             The next issue that you have to deal with,

 

 14   if you have got cells and you grow them and you

 

 15   inject them into an animal, is how you measure the

 

 16   outcome, and the question of safety is obviously an

 

 17   open one, and none of us really have addressed the

 

 18   safety question in preclinical models.

 

 19             We didn't really know that there was going

 

 20   to be a safety issue.  I was talking at lunch about

 

 21   the fact that, you know, we had a number of animals

 

 22   drop dead over the course of our studies when we

 

 23   were doing these experiments early on, but we

 

 24   assumed it was because we were doing open-chest

 

 25   surgeries on these animals to create the infarct,

 

                                                               178

 

  1   to deliver the cells, to measure cardiac function,

 

  2   not that it could have anything to do with a

 

  3   potential electrical effect of the cells in vivo.

 

  4             So, we had to then go back and evaluate

 

  5   whether or not safety was even at all compromised

 

  6   or relevant in these animal models, so none of us

 

  7   are really measuring safety. There have been two

 

  8   studies reported, one in a pig model of holter

 

  9   monitoring animals, and we just presented some data

 

 10   at ACC in rabbit, monitoring electrical effects of

 

 11   myoblasts, and I will show some of those data in a

 

 12   minute.

 

 13             The other issue in terms of measuring

 

 14   outcome is function.  Typically, in rodents,

 

 15   isolated heart preps have been used to measure

 

 16   function although in some cases, sonomicrometry is

 

 17   used.  I have actually put the methods here in

 

 18   order of I believe their ability to actually give

 

 19   you useful information.

 

 20             I think the isolated heart prep is the

 

 21   least useful because it is subject to a lot of

 

 22   variability, it is subject to baseline drift, it is

 

 23   subject to flow and rate factors, and it is subject

 

 24   to ischemia in vitro.

 

 25             Echocardiography is obviously used

 

                                                               179

 

  1   clinically, as well as experimentally, but it

 

  2   varies dramatically with the operator and the

 

  3   orientation of the probes, so you can make echo

 

  4   tell you just about anything you want.

 

  5             If you really want to measure work done in

 

  6   a region of the heart, I submit that you have to go

 

  7   back and do sonomicrometry in that area and use

 

  8   crystals to actually measure the ability of that

 

  9   region of the myocardium to move in an electrically

 

 10   and mechanically meaningful way.

 

 11             Then, more recently, cine MRI has really

 

 12   come to the fore in terms of our ability to make

 

 13   measurements in not only people, but animals, as

 

 14   well.

 

 15             But measuring function is pretty useless

 

 16   unless it correlates with histology, and we begin

 

 17   to ask questions about angiogenesis and myogenesis,

 

 18   and we don't always correlate histology with

 

 19   outcome, and, in fact, one of the issues that comes

 

 20   up over and over is clinically, as well as

 

 21   experimentally, there is a disconnect between the

 

 22   number of cells we can find in the heart and the

 

 23   functional improvement we see, which begins to ask

 

 24   questions about mechanism.

 

 25             So, in terms of myoblasts, what are these

 

                                                               180

 

  1   cells? As I said, there are myoblasts or

 

  2   fibroblasts and there are SP cells.  I propose that

 

  3   the mechanism of repair of these cells depends on

 

  4   the number of cell types that you have present and

 

  5   the percentage of each.

 

  6             I believe myoblasts know to become muscle,

 

  7   and they are capable of myogenesis.  I believe the

 

  8   fibroblasts not only secrete an angiogenic factor

 

  9   FGF, but also act as a growth factor and a mitogen

 

 10   for myoblasts.

 

 11             That has been known for years.  Judy

 

 12   Swain's data, they actually keep myoblasts alive

 

 13   and keep myoblasts proliferating over a fairly

 

 14   extended period of time, FGF does.

 

 15             SP cells, I believe are more likely to be

 

 16   angiogenic and also possibly to fuse with other

 

 17   cells in the myocardium.  That is based on data

 

 18   primarily from our group showing that the more

 

 19   immature a cell is, the more likely it is to fuse.

 

 20             So, in terms of our animal models, I think

 

 21   the question we have to ask is what do the patients

 

 22   look like first, and in the myoblast trials, those

 

 23   patients have been post infarction, usually greater

 

 24   than one month, up to many  years.  The average in

 

 25   one of the studies was 6.7 years.

 

                                                               181

 

  1             Most of those patients are in need of

 

  2   revascularization, they have an ejection fraction

 

  3   of less than 35 percent, and they have heart

 

  4   failure.

 

  5             If you look at the European experience,

 

  6   Philippe has already talked about some of this, and

 

  7   I am not going to go over these in detail.  This is

 

  8   a slide that was given to me by Peter Smits from

 

  9   Rotterdam.

 

 10             You look at Spain and Poland, and

 

 11   obviously the French study, and then the U.S.

 

 12   studies, both Arizona heart and Bioheart Mount

 

 13   Sinai study, and then the Bioheart study in Europe,

 

 14   and I should say for the sake of disclosure that I

 

 15   have had a relationship with Bioheart, so take

 

 16   these data with a grain of salt, that all of these

 

 17   patients are heart failure patients.

 

 18             The average in this study is 6.7 years

 

 19   post-infarction, so these are patients who are

 

 20   already pretty sick, and who have significant

 

 21   electrical abnormalities already.

 

 22             How do the preclinical patients compare?

 

 23   Well, for myoblast studies, these animals all have

 

 24   acute cardiac injury, either, as I said, cryoinjury

 

 25   or coronary artery ligation.  Very few, if any,

 

                                                               182

 

  1   studies deal with occlusion reperfusion, which is

 

  2   what happens clinically.

 

  3             You can open up virtually any artery in

 

  4   the heart now, but nobody is doing preclinical

 

  5   studies where we do ischemia reperfusion.  We are

 

  6   just now moving in that direction.

 

  7             I think the reason initially for at least

 

  8   us, and I believe for other people, was that we

 

  9   wanted to kill everything that was there, so that

 

 10   anything we found was due to something we put in,

 

 11   and so we started with cryoinjury where we applied

 

 12   a minus 70 degrees C probe to the surface of the

 

 13   heart to wipe out that region of the heart, but it

 

 14   raises questions about the inflammatory process,

 

 15   which we are coming to understand is critical in

 

 16   terms of potential homing of cells and the

 

 17   potential mechanism.

 

 18             Most of the preclinical studies, the cells

 

 19   are delivered two to three weeks post-injury, and

 

 20   there is a one- to three-month follow-up.

 

 21   Clinically, this isn't exactly relevant.

 

 22             I just told you that every clinical study

 

 23   is at least one month post-injury and sometimes six

 

 24   to seven years post-injury, so we are not looking

 

 25   at the same milieu into which we put these cells.

 

                                                               183

 

  1             In term of heart failure models, there are

 

  2   a few models out there.  Dan Burkoff's group has

 

  3   recently published a study in dog where they used

 

  4   microspheres to actually create a heart failure

 

  5   model, and have gotten data with myoblasts that

 

  6   actually look very similar to some of the data

 

  7   gathered in earlier models.

 

  8             None of the studies to date have really

 

  9   used any animals with LVAD support, and yet there

 

 10   are clinical trials beginning to move forward in

 

 11   that, and actually, one has already been completed

 

 12   in that context.  So, we would expect that with a

 

 13   completely unloaded heart, we might have very

 

 14   different phenomena.

 

 15             In terms of cardiomyopathy, there is a

 

 16   hamster model and mouse genetic models that have

 

 17   begun to be used for myoblast transplantation, and

 

 18   Magdia Koob's group has actually published

 

 19   reasonable data in terms of a rat model of

 

 20   adriamycin toxicity.  So, we are beginning to get a

 

 21   plethora of models in which we can look at myoblast

 

 22   transplantation.

 

 23             I put up here actually the little bit of

 

 24   physiology that I could pull together about some of

 

 25   the different animal models just to make a few

 

                                                               184

 

  1   points.

 

  2             One is that when you start looking at

 

  3   these different injury models in mouse, rat,

 

  4   rabbit, dog, pig, sheep, and humans, that there

 

  5   really are significant differences in rodents and

 

  6   the larger animals.

 

  7             Mouse and rat, you know, the hearts are

 

  8   pretty darn small.  Their heart rate consequently

 

  9   is very high. These animals have very few

 

 10   collaterals, and they express completely different

 

 11   contractile proteins than are expressed in the

 

 12   majority of the heart.

 

 13             Moreover, the action potential in the

 

 14   electrical capacity of the mice and rat is very

 

 15   different.  There is no plateau phase in the action

 

 16   potential in mice, and the action potential

 

 17   duration is on the order of 10 milliseconds.  In

 

 18   humans, it is on the order of 250 milliseconds.

 

 19             Rabbit is the first animal model where you

 

 20   begin to get numbers and conditions that resemble

 

 21   humans, and that is why we chose rabbit early on,

 

 22   and I would submit that in terms of feasibility

 

 23   studies, rabbit is a good entry level animal under

 

 24   most conditions except where you are trying to do

 

 25   stem cell work.

 

                                                               185

 

  1             When you are trying to do stem cell work,

 

  2   we don't have the markers for stem cells in most of

 

  3   these other species that we do in rat, mouse, and

 

  4   humans, so that is when mice, rats, and humans, or

 

  5   maybe pigs, some of the human cytokines and

 

  6   antibodies cross to pig, but not all of them, so

 

  7   you can begin to do some of those studies in pig.

 

  8             Nonetheless, in terms of feasibility

 

  9   studies, I think rabbit is a good model, and you

 

 10   move up from there.

 

 11             So, let's look at each of these.  In terms

 

 12   of myoblast in the mouse, you can begin to track

 

 13   the cells because you can use genetic models of

 

 14   where the cells actually express different markers

 

 15   that are unavailable in the animals in which you

 

 16   inject the cells.

 

 17             You can begin to isolate and characterize

 

 18   stem cells including the stem cells in muscle, but

 

 19   you can't characterize from larger species.  You

 

 20   can use immunocompromised mice for human cells, but

 

 21   you are missing an important component, which is

 

 22   the inflammatory component, and we are beginning to

 

 23   understand again relates to homing and perhaps even

 

 24   recruitment of cells.

 

 25             What is the advantage of a rat versus a

 

                                                               186

 

  1   mouse? Well, the main advantage is it is larger

 

  2   than a mouse, so you can do a few more things, but

 

  3   you can still track the cells and isolate stem

 

  4   cells, and you can still have an immunocompromised

 

  5   model.

 

  6             The rabbit, the bottom line about rabbits

 

  7   is most people make antibodies in rabbits, not

 

  8   against rabbits, so it is really hard to find the

 

  9   tools that you need to do some of the evaluations

 

 10   downstream, but it is still a relatively

 

 11   inexpensive model with cardiac characteristics very

 

 12   similar to humans.

 

 13             The pig, obviously, the size is good, and

 

 14   the geometry is good for catheter-based studies.

 

 15   One of the points I want to make about delivery of

 

 16   these cells, and I think Philippe showed it when he

 

 17   was talking about his clinical trials, and we have

 

 18   done the same thing in terms of our preclinical

 

 19   studies, surgical studies, is that when you inject

 

 20   these cells surgically, most of us have delivered

 

 21   the cells parallel to the surface of the heart.

 

 22             We have done that for years because we

 

 23   really thought it was going to increase the number

 

 24   of cells that we could get into the myocardium.

 

 25   Yet, all of the catheter-based studies deliver the

 

                                                               187

 

  1   cells perpendicular to the surface of the heart,

 

  2   and it is not completely unexpected that geometry

 

  3   may make a difference in terms of how these cells

 

  4   actually function in the myocardium.

 

  5             So, I think it is important to evaluate

 

  6   the geometry of the cells in some of these larger

 

  7   animal models.

 

  8             So, how do you choose an animal model for

 

  9   the myoblast studies?  I think feasibility and

 

 10   costs are obviously important, whether or not you

 

 11   are going to do high throughput studies and need to

 

 12   track your cells.  Rodent and  hamster I think are

 

 13   best for those.  Rabbit is best in terms of

 

 14   beginning to be physiologically relevant to humans

 

 15   in terms of heart rate and scalability.  I will

 

 16   show some data in a minute in terms of scalability.

 

 17             The large animal models are obviously much

 

 18   more physiologically relevant.  You can get a sense

 

 19   of dose.  You can use conventional delivery methods

 

 20   that you would use in humans.  You can do the right

 

 21   functional assessments, and the heart size and

 

 22   geometry is very similar to a human.

 

 23             So, what exists for myoblasts?  Well, as I

 

 24   said, the route of administration has been

 

 25   primarily surgical or percutaneous.  Intravenous

 

                                                               188

 

  1   and intracoronary studies are just beginning

 

  2   although we published some intracoronary data, that

 

  3   was the first thing we published in '96.

 

  4             Myoblasts are unlike stem cells or unlike

 

  5   bone marrow mononuclear cells.  They are very much

 

  6   like stromal cells.  They are big.  When we put

 

  7   myoblasts in the coronary circulation, what we

 

  8   found is that we got profound ST elevations, and we

 

  9   saw transient ischemia every time we injected these

 

 10   cells.

 

 11             So, we actually think that with large

 

 12   cells, that the way they are actually having an

 

 13   effect in the myocardium is creating essentially a

 

 14   microinfarct clogging the vessels and then getting

 

 15   out of the vessels as a result of that.

 

 16             Mononuclear cells are much smaller, and I

 

 17   think don't have the same effect.

 

 18             In terms of dose, in a mouse, typically,

 

 19   you give about 1,000 cells.  Some people go as high

 

 20   as a million, but typically, 1,000 is enough to

 

 21   begin to see an effect.

 

 22             In our hands, in rabbit, the lowest dose

 

 23   at which we see an effect is 3 x 10                                      

                                                  7 cells.  We

 

 24   tried 10                                            7, 3 x 107, 108, and

3 x 108, and this is the

 

 25   range in which we see the most effect.

 

                                                               189

 

  1             In pig, it is about 3 x 10                                     

                                                     8, and what we

 

  2   found, you know, pig is about 10 times bigger than

 

  3   a rabbit, you need about 10 times as many cells.

 

  4   Rabbit is about I think about 4,000 times the size

 

  5   of a mouse, and we found that we need many more

 

  6   than 4,000 more cells in a rabbit than we do in a

 

  7   mouse.  So, I don't think you can really

 

  8   extrapolate from mouse, but I think you can begin

 

  9   to extrapolate at the size of rabbit and go up.

 

 10             In terms of cell location and where

 

 11   myoblasts have been injected, you pick a surgical

 

 12   fellow who is doing the experiments, and you will

 

 13   get a different location of injection virtually

 

 14   every time, I guarantee it, and you are not going

 

 15   to convince them otherwise that their way isn't the

 

 16   right way to do it.  It is completely ignored in

 

 17   most of the preclinical studies.

 

 18             There might be mention of one injection or

 

 19   two injections or three injections, but in terms of

 

 20   the exact location, I couldn't tell you, I couldn't

 

 21   find in the literature where the majority of

 

 22   injections occur.  I know in my own lab, it is not

 

 23   consistent from study to study.

 

 24             In terms of timing, myoblasts have been

 

 25   injected   two to four weeks post-injury.  The

 

                                                               190

 

  1   vehicle has been PBS cell growth medium minus

 

  2   serum, or it is completely ignored, there is no

 

  3   mention of it.

 

  4             So, this is the slide again I borrowed

 

  5   from Philippe's work to illustrate how he did some

 

  6   of the early injections with a bent needle again

 

  7   parallel to the surface of the heart, and also that

 

  8   the injections are done, not just in the center of

 

  9   the infarct, but in the periinfarct region, as

 

 10   well.

 

 11             Similarly, with a percutaneous approach,

 

 12   and this is another slide from Peter Smits, in the

 

 13   first patient who received cells, and in the

 

 14   majority of cases now with percutaneous myoblast

 

 15   delivery, cells are delivered in the periinfarct

 

 16   region, in the normal region of myocardium, and

 

 17   very few of the injections percentagewise actually

 

 18   end up in the infarcted cell, and that may have an

 

 19   effect on safety, and I will show some preclinical

 

 20   data that support that.

 

 21             So, the majority of injections are

 

 22   surgical, and you can inject the cells and find

 

 23   them in the center of the infarct.  What you get

 

 24   surgically when you inject these cells is one or

 

 25   two things.

 

                                                               191

 

  1             On a great day when you are really lucky,

 

  2   you get what looks like a chunk of steak in the

 

  3   center of the heart.  On a typical day, you see

 

  4   something that looks more like this, where you have

 

  5   patchy regions of cells distributed through the

 

  6   infarct rather than these large fibers that you see

 

  7   here, and these patchy cells distributed throughout

 

  8   the infarct are not necessarily talking to each

 

  9   other, but they are all oriented with the

 

 10   extracellular matrix.

 

 11             You can see there are some small vessels

 

 12   here, here.  We often see large vessels in the

 

 13   infarct, as well.

 

 14             These are preclinical data from my group,

 

 15   but they don't look too dissimilar from what you

 

 16   see from Pagani's paper from myoblasts in an

 

 17   LVAD-supported human heart or, in fact, the data

 

 18   that Philippe showed you earlier of the myoblasts

 

 19   surrounded by scar in a patient 17 1/2 months after

 

 20   injection.

 

 21             I am not going to talk about stromal cells

 

 22   because that is not my job today, but what I am

 

 23   going to begin to talk about is delivery.

 

 24   Assessing delivery requires that we be able to

 

 25   track the cells.  In vivo, we have chosen SPECT or

 

                                                               192

 

  1   MRI most recently, although I think PET is going to

 

  2   be a good method, as well.

 

  3             That has to correlate in vitro with

 

  4   histology and appropriate markers.  If I don't make

 

  5   any other point today, take home the fact that

 

  6   using desmin, using phospholamban, using GATA-4,

 

  7   using all of these markers that people claim are

 

  8   cardiac markers, are not cardiac-specific markers.

 

  9   You find these markers in other muscle cells, you

 

 10   find these markers in undifferentiated progenitor

 

 11   cells.

 

 12             If you look in C2C12 skeletal muscle

 

 13   cells, you can see phospholamban, you can see

 

 14   connexin 43, you can see in some cases, in

 

 15   progenitor cells, you see GATA-4.  You have got to

 

 16   use markers that are specific for cardiocytes if

 

 17   you are going to call these cells cardiac cells,

 

 18   and the only markers that I know of right now, that

 

 19   I believe are specific for cardiocytes, are channel

 

 20   markers that are actually not expressed in skeletal

 

 21   muscle.

 

 22             As skeletal muscle matures, it expresses

 

 23   many cardiac-specific proteins, and as skeletal

 

 24   muscle matures, it expresses cardiac markers, as

 

 25   cardiac muscle matures, it expresses skeletal

 

                                                               193

 

  1   markers.  So, we don't know where in that process

 

  2   we are, so we can't really use those markers.

 

  3             This is an image showing that we can begin

 

  4   to visualize these cells in the heart.  These are

 

  5   indium-111 labeled myoblasts present in a short

 

  6   axis view by SPECT imaging of a rabbit heart

 

  7   showing that we can actually co-deliver

 

  8   tetrofosmin, see perfusion, see the dropoff in

 

  9   perfusion here with the infarct, and then see the

 

 10   indium-labeled cells in the center of the image.

 

 11             So, we are beginning to believe that we

 

 12   can actually track cells over time now.  This is

 

 13   also a cine MRI of a rabbit heart, and these are

 

 14   data that were all gathered at Duke.  This is a

 

 15   rabbit heart, so at the level of rabbit, although

 

 16   we can now do the same thing in a mouse, we can

 

 17   iron label our cells in a way that we believe

 

 18   doesn't affect proliferation or viability of the

 

 19   cells, and begin to see them in the center of the

 

 20   infarct region, the infarct region here being

 

 21   contrast-enhanced in white.

 

 22             So, we can start now to label these cells.

 

 23   We have followed these cells out to four months in

 

 24   this way and can still find them.  When we kill the

 

 25   cells and then inject them, the iron label goes

 

                                                               194

 

  1   away over about two to three days, so we are fairly

 

  2   convinced that the iron is present in viable cells.

 

  3             So, the other issues in terms of measuring

 

  4   outcome, I think I have already made this point,

 

  5   safety is an open question, and I think what I take

 

  6   away from the field so far is if you don't look,

 

  7   you won't find it, and that we didn't look, and now

 

  8   I think it is important that we begin to do holter

 

  9   monitoring and other electrically relevant studies,

 

 10   and those are going to require large animals, pig,

 

 11   rabbit.  You can't do those in mouse and rat, not

 

 12   at 300 to 600 beats per minute.  You are really not

 

 13   going to be able to see a VF or a VT.

 

 14             In terms of function, I think if the goal

 

 15   here with myoblasts is really to find an

 

 16   ischemia-resistant cell that is electrically

 

 17   compatible with a healthy heart, we have got to

 

 18   also look at electrical activity of these cells

 

 19   over time.

 

 20             This is again a slide that Peter Smits

 

 21   provided showing clinical data and the number of

 

 22   VPCs per visit in some of the early patients who

 

 23   had cells delivered, and I modified the slide a bit

 

 24   to show times at which patients have actually died

 

 25   after cell delivery.

 

                                                               195

 

  1             What you begin to see is that there is a

 

  2   window of time from about a week to a month where

 

  3   there seems to be an increased incidence of

 

  4   electrical abnormalities.  When we have done animal

 

  5   studies now, we see that same sort of window from

 

  6   about 3 days to about 3 1/2  weeks, and then it

 

  7   drops off and we don't see the incidence after that

 

  8   period of time.

 

  9             So, the safety may depend on the cell

 

 10   dose.  We have found that if we just look at PVCs

 

 11   in our animal models, that as we increase dose, we

 

 12   increase the number of PVCs, and it may also depend

 

 13   on location.

 

 14             We have found that if we inject cells in

 

 15   the center of the infarct and we measure PVCs, and

 

 16   this is actually 10                                                      

       8 cells, 107 is not functionally

 

 17   relevant, 10                                                   8 is, 109

is, we found that if we inject

 

 18   cells in the center of the infarct, we see PVCs and

 

 19   no monomorphic VT.

 

 20             If we begin to inject cells in the border

 

 21   zone, we not only increase the number of PVCs we

 

 22   see, but we start seeing runs of ventricular

 

 23   tachycardia.  If we inject cells in both the center

 

 24   and the periphery, we see essentially the same

 

 25   thing, and more up-to-data were just presented at

 

                                                               196

 

  1   the ACC from my lab.

 

  2             What is interesting is we began to take

 

  3   these cells back out of heart, what we found is

 

  4   that their action potential duration changed, that

 

  5   initially, the cells had an action potential

 

  6   duration of on the order of 20 seconds, and over

 

  7   time it increased to something on the order of 120

 

  8   milliseconds, but it is still not compatible with

 

  9   the surrounding heart.

 

 10             We have also done some modeling data.

 

 11   What we believe is that if you have these cells

 

 12   coupled to each other, that is a good thing in the

 

 13   center of the infarct, but that you don't want them

 

 14   coupled to the remainder of the heart until they

 

 15   are electrically compatible with the remainder of

 

 16   the heart, and yet clinically, very little

 

 17   attention has been paid to location.

 

 18             Again, it is an issue that we didn't know

 

 19   we were going to have to address, and now we have

 

 20   got to go back and address.  In fact, some of the

 

 21   locations of injections could explain why there

 

 22   have been ventricular tachycardia in some of these

 

 23   patients.

 

 24             The only possibility is, you know, over

 

 25   that window of time, we don't know if these cells

 

                                                               197

 

  1   are integrating, dying, or changing their

 

  2   phenotype, we have no idea, and I think we really

 

  3   have to begin to elucidate that.

 

  4             So, in a standardized model where we know

 

  5   how myoblasts function, we have now got to look at

 

  6   location, dose, and route of administration.

 

  7             I think I have already said this, so I am

 

  8   not going to really belittle, spend time on

 

  9   function especially  other than to say we have

 

 10   begun to collect a lot of data now with a lot of

 

 11   different cell types and a lot of different  growth

 

 12   factors, and what we have begun to realize is that

 

 13   virtually anything we put into the heart, cells,

 

 14   myoblasts, fibroblasts, bone marrow stromal cells,

 

 15   bone marrow mononuclear cells, growth factors

 

 16   including VEGF and other growth factors, improve

 

 17   the mechanical properties of the scar, and change

 

 18   diastolic performance.

 

 19             They do that first, before they have any

 

 20   effect on  systolic performance, usually by several

 

 21   weeks.  What we figure is that having something

 

 22   alive in the scar is better than having just this

 

 23   dense collagen matrix, and it really doesn't seem

 

 24   to matter what you have alive in the scar, if is

 

 25   vessels, if it is muscle, if it is whatever, you

 

                                                               198

 

  1   improve compliance.

 

  2             But we don't see the corresponding

 

  3   improvement in systolic performance, at least not

 

  4   with fibroblasts in our hand, but we do with

 

  5   myoblasts, we don't with VEGF, but we have now with

 

  6   bone marrow stromal cells and bone marrow

 

  7   mononuclear cells.

 

  8             These are some data that just came out in

 

  9   Circulation showing that, that if we use crystals

 

 10   to measure regional stroke work in our sham-treated

 

 11   animals, regional function gets worse, but in our

 

 12   myoblast-treated animals, function goes from pretty

 

 13   bad to better, and in our bone marrow stromal cell

 

 14   animals, the same thing is true.

 

 15             I think that really raises a question

 

 16   about mechanism, but the positive outcome in our

 

 17   hands at least is dose dependent, 10                                     

                                                     7 no effect, 108

 

 18   positive effect, sham continues to get worse.

 

 19             What is interesting is this is not just

 

 20   improvement versus cell number.  This is log of

 

 21   injected cells, but this is the percentage of

 

 22   animals that actually improve.

 

 23             So, what we found is that the percentage

 

 24   of animals increases with cell dose, as well.

 

 25             So, will myoblast transfer work in

 

                                                               199

 

  1   patients? Philippe already has told us that it

 

  2   will, and we have begun to believe that, in fact,

 

  3   that there are different mechanisms of action for

 

  4   these cells.

 

  5             I think I will just very quickly go

 

  6   through the last couple slides.  We believe that

 

  7   myoblasts improve both regional and global function

 

  8   in the heart based on our preclinical data.

 

  9             If we use cine MRI and actually measure

 

 10   thickness in the wall of the myocardium over time

 

 11   and global wall thickening, so areas where cells

 

 12   were not injected, we use contrast to define where

 

 13   the infarct is, and this is area that has no

 

 14   contrast in it, so the remainder of the heart

 

 15   actually gets better, wall thickening improves in

 

 16   the cell treated, but not in the control vehicle

 

 17   injected animals.

 

 18             Regional wall thickening where we actually

 

 19   inject the cells gets better to a greater degree,

 

 20   so we only measured this where there was a

 

 21   transmural infarct.  We didn't measure it in

 

 22   regions at the periphery of the infarct where there

 

 23   can be tethering going on.  So, we use contrast and

 

 24   only measured it in the region.

 

 25             Diastolic volume decreased, heart weight

 

                                                               200

 

  1   decreased, so global indices of failure also

 

  2   improved.

 

  3             Every cell we and virtually anyone has

 

  4   injected seems to work, which either means the

 

  5   myocardium is easier to repair than we thought or

 

  6   we don't understand what is happening and we aren't

 

  7   looking at the data correctly.

 

  8             I would like to believe it is this, and I

 

  9   am actually going to posit this in a little bit,

 

 10   but I have a bad feeling.  I would also submit that

 

 11   they work despite the fact that we don't know how

 

 12   to get the cells there in large numbers, and we

 

 13   can't always find them histologically, and that we

 

 14   don't really know what to look for.

 

 15             These cells may be promoting angiogenesis,

 

 16   myogenesis, they may just be unloading the heart,

 

 17   changing wall stress.  They may be secreting

 

 18   paracrine factors that recruit other endogenous

 

 19   stem cells to the area of injury, either cells from

 

 20   the heart, if that's your fancy, of cells from the

 

 21   bone marrow, or maybe a combination thereof, or

 

 22   maybe they work because we are lacking long-term

 

 23   follow-up in both animals and patients, and we

 

 24   haven't asked the right questions.

 

 25             So, I will just stop by saying we have

 

                                                               201

 

  1   started in small animals, we have moved to large

 

  2   animals, and we have moved to patients, and these

 

  3   are the cells that we knew the most about, and we

 

  4   have had to say you know what, we missed a lot, and

 

  5   we have had to go back and take the safety and

 

  6   functional effects that we have seen and reevaluate

 

  7   them in all of these animal models again, and I

 

  8   think that should be a lesson for going forward.

 

  9             Is it time for randomized trials, and, if

 

 10   so, who? The think the questions we have to ask is,

 

 11   is it safe, who are the right patients, what are

 

 12   the appropriate endpoints.

 

 13             I show this every time.  If we do this

 

 14   wrong, we are really going to doom what I think is

 

 15   an exciting field. I don't want to be standing up

 

 16   here talking to you about the gene therapy lessons

 

 17   that we could have learned and didn't.

 

 18             I think what we can learn from gene

 

 19   therapy is in 6 open-label trials, they were all

 

 20   positive, and 4 out of 5 randomized, double-blind,

 

 21   control trials, they weren't.  Is that because

 

 22   patients got better care, or because there was a

 

 23   better placebo effect?  Well, we won't really know

 

 24   until we do the randomized trials, but we need to

 

 25   under-promise and over-deliver, not conversely.

 

                                                               202

 

  1             These are the people who did all the work,

 

  2   and I thank you.

 

  3             [Applause.]

 

  4             DR. RAO:  Thank you, Doris.

 

  5             We are open for questions.

 

  6                               Q&A

 

  7             DR. ROSE:  Dr. Taylor, I enjoyed your

 

  8   talk, but I must say, as a clinical cardiologist, I

 

  9   just have difficulty in envisioning how these

 

 10   myotubules, that presumably result from the

 

 11   injection of the myoblasts into scar, increase

 

 12   systolic wall thickening, which is what I would

 

 13   accept as evidence of improved regional function.

 

 14             Unfortunately, many of your images weren't

 

 15   showable, but was the increase in thickening that

 

 16   you showed on that on that one graphic slide, was

 

 17   that from your larger animal models as opposed to

 

 18   your smaller animal models?

 

 19             DR. TAYLOR:  No, that was from rabbit.

 

 20             DR. ROSE:  You mentioned early in your

 

 21   talk about a disconnect between the numbers of

 

 22   cells and the functional improvement.  Is that true

 

 23   for the myoblast injection?  In other words, there

 

 24   was probably a range of responses, I would imagine,

 

 25   some had perhaps better systolic thickening

 

                                                               203

 

  1   improvement than others, and by histologic

 

  2   examination, were there more myotubules or were the

 

  3   myotubules oriented in a different direction than

 

  4   the animals that perhaps had less improvement in

 

  5   systolic thickening?

 

  6             DR. TAYLOR:  Have we quantified that

 

  7   unequivocally?  No, because it is very difficult to

 

  8   quantify the number of cells in an infarct and know

 

  9   that they are actually myoblasts in the cells you

 

 10   injected because we are giving autologous cells.

 

 11             One of the rate limiting steps in this

 

 12   field is having good markers for the cells, and

 

 13   that is one of the reasons we went to iron, so that

 

 14   we could actually stain our sections later for

 

 15   Prussian blue and look for iron and say, okay,

 

 16   these are the cells we injected.  We shouldn't see

 

 17   that in normal cells.

 

 18             So, we are starting to now look and try to

 

 19   answer that question.  What we do know is that with

 

 20   a number of studies that have been done by a number

 

 21   of groups, the number of cells that you retain in

 

 22   the scar after you inject them is about 15 to 25

 

 23   percent of the cells you initially put in there

 

 24   probably.  On a good day, maybe 30, 40 percent of

 

 25   the cells you put in there, but that is rare.

 

                                                               204

 

  1             What we also know is that numbers of cells

 

  2   can die over time, but we also know that these

 

  3   cells can proliferate over time, and when we look

 

  4   for proliferation, we in fact can find that in

 

  5   these scarred regions.

 

  6             I don't actually think that we are seeing

 

  7   evidence of long myofibers necessarily.  We see

 

  8   cells that line up with each other and connect with

 

  9   each other, and we believe that these cells are

 

 10   being mechanically induced to contract.

 

 11             We know when you stretch muscle, it

 

 12   contracts.  Do I think they are electrically

 

 13   coupled with the rest of the heart?  No.  Do I

 

 14   think they are mechanically coupled?  Yes. What we

 

 15   see is we measure left ventricular pressure going

 

 16   up, we see pressure plateau, and right at the time

 

 17   that pressure within a few milliseconds after

 

 18   pressure plateaus, we see our crystals move in that

 

 19   region of the heart, and then pressure decreases,

 

 20   and then we see that again.

 

 21             We can track that beat after beat after

 

 22   beat in the scarred region.  We can go from

 

 23   negative work loops where the rest of the heart is

 

 24   being pushed by the remainder of the heart to

 

 25   positive work loops when we put the

 

                                                               205

 

  1   cells--actually, I have got it backwards--negative

 

  2   in this direction versus positive work loops, that

 

  3   actually correspond to systole.

 

  4             So, I believe that there is wall

 

  5   thickening and actual contraction going on in that

 

  6   region, and I think our sonomicrometry data are the

 

  7   most convincing data.

 

  8             Is it possible to get the image to show,

 

  9   so I could try to show one of the functional wall

 

 10   thickening images, so you can actually see that?

 

 11   If you just give me the slide thing back, I will

 

 12   try.

 

 13             I am going to try this first.

 

 14             So, this is, in theory, it is beating

 

 15   here.  No, apparently I can't, maybe you can, but

 

 16   we are able to actually show thickening and I will

 

 17   pull my computer out to show anybody later who

 

 18   wants to see it.

 

 19             You can actually see in the sham-treated

 

 20   animal, here is the scarred region and only in the

 

 21   very center of the scar is it not thickening

 

 22   anymore.  The scar was from about here to here, by

 

 23   contrast.

 

 24             The scar of the sham-injected animals over

 

 25   here is not thickening at all.  Here, we only don't

 

                                                               206

 

  1   see thickening right in the very center of the

 

  2   scar, and we used contrast enhancement, so we took

 

  3   10 slices through the rabbit heart, in a long axis

 

  4   view, we used contrast, and only where we saw

 

  5   contrast gadolinium did we call that infarct, and

 

  6   then measured thickening in that region, and we did

 

  7   that in a blinded way.

 

  8             We are pretty convinced that we see wall

 

  9   thickening in that region.

 

 10             DR. BLAZAR:  You made a good point that

 

 11   the larger animals allow you to assess function

 

 12   much more directly and extrapolate to humans.  With

 

 13   the smaller animal models, you have a higher

 

 14   throughput.  So, the question is, what is the data

 

 15   that says that the smaller animals extrapolate, the

 

 16   large animals extrapolate to the human as you go

 

 17   through all of these different examples, because

 

 18   one would hear your presentation and think that it

 

 19   really just should be restricted to large animals

 

 20   minus a few, more esoteric.

 

 21             DR. TAYLOR:  What I can say is we have

 

 22   made measurements in rabbit and pig for many, many

 

 23   years, and I swore we would never use mice or rats,

 

 24   and then we decided that we wanted to start making

 

 25   comparisons with bone marrow derived cells, and we

 

                                                               207

 

  1   didn't have the markers to isolate those cells in

 

  2   rabbit or pig, so we went back and did the same

 

  3   experiments in mice, and we got the same results.

 

  4             So, now I have to bite my tongue and show

 

  5   mice data even though I swore we never would.  I am

 

  6   convinced at least with MRI, that we can make

 

  7   meaningful measurements that show us the same sort

 

  8   of thing.

 

  9             DR. BLAZAR:  Although you also made a

 

 10   point as to the dose, location, et cetera, it would

 

 11   seem that that is going to be extremely difficult.

 

 12             DR. TAYLOR:  It is very difficult.  That

 

 13   being said, what we do is we inject the cells in

 

 14   the center of the scar in a mouse.  Do I believe we

 

 15   get the same percentage that we get in larger

 

 16   animals?  No.  We really don't know  what number we

 

 17   actually get in.

 

 18             We have started doing some biodistribution

 

 19   studies to try to answer that, and we are mostly

 

 20   doing those in rabbit and pig, because I have no

 

 21   confidence for the numbers we get in mice.  In

 

 22   terms of doing stem cell studies, though, I think

 

 23   it is critical that you use something where you can

 

 24   clearly define the cells.

 

 25             DR. ALLAN:  A little bit of a follow-up,

 

                                                               208

 

  1   which is use of nonhuman primates.  Somebody

 

  2   mentioned that there were some primate studies.

 

  3   This morning, I think somebody just referred to

 

  4   them.

 

  5             DR. TAYLOR:  Right, with bone marrow

 

  6   cells.

 

  7             DR. ALLAN:  Is it with bone marrow cells?

 

  8   Because you can use those markers on many of the

 

  9   nonhuman primates,  you can look at stem cells.

 

 10             DR. TAYLOR:  Right.

 

 11             DR. ALLAN:  Maybe to use that model as a

 

 12   step between "large" animals and humans, because

 

 13   anything that you have derived that looks promising

 

 14   could through that nonhuman primate.

 

 15             DR. TAYLOR:  I think that is good point,

 

 16   and we have actually started collaborating with

 

 17   some people in California who do nonhuman primate

 

 18   studies.  I can tell you that they are God-awful

 

 19   expensive, they are hard to do, and I personally

 

 20   find them hard to do.

 

 21             Sometimes we just have to get over it, but

 

 22   I think if the pig data are good enough, I would

 

 23   rather stick with pigs, and the fact that we can

 

 24   use some of the same stem cell markers has made me

 

 25   focus more in that area.

 

                                                               209

 

  1             DR. BORER:  That was a very interesting

 

  2   presentation.  I have a question that sort of falls

 

  3   into are we looking at the data the right way or

 

  4   oversimplifying box.  You can't argue with success,

 

  5   and I believe that some contractility is probably

 

  6   better than done and perfect is the enemy of good,

 

  7   and all that stuff, but, you know, you have put in

 

  8   skeletal muscle cells, and they contract, and yet

 

  9   to get useful work from the heart, forced

 

 10   generation is just part of the equation, you have

 

 11   to have forced transmission, as well, and I haven't

 

 12   heard anything yet about remodeling at the

 

 13   extracellular matrix and regeneration of dystrophin

 

 14   ECM hookups, and whatever, that might demonstrate

 

 15   that we are actually developing a forced

 

 16   generation/forced transmission system.

 

 17             I don't know how important that is

 

 18   ultimately, but it makes me wonder, the fact that

 

 19   we haven't heard about that, and I am not sure that

 

 20   I could expect it would happen, that we are

 

 21   actually looking at the data the right way, so

 

 22   could you talk a little bit about that?

 

 23             DR. TAYLOR:  Sure.  The one piece of data

 

 24   that we do have, that I think addresses that, is

 

 25   the compliance data where we looked at changes in

 

                                                               210

 

  1   strain with the different cell types, and that

 

  2   begins to address matrix and what is going on in

 

  3   that matrix - does it look at signaling, does it

 

  4   look at MMPs, does it look at any of that?  No.

 

  5             I think the bottom line is when we started

 

  6   this 15 years ago, our goal was to show it worked,

 

  7   and then once we showed it worked, we thought we

 

  8   would go back and figure out how it worked.

 

  9             As soon as we showed it worked, God and

 

 10   everybody wanted to do it clinically, and so we are

 

 11   having this discussion rather than understanding

 

 12   how it worked, which we have had to develop in

 

 13   parallel.  I think we are still catching up in

 

 14   terms of trying to develop, trying to understand

 

 15   the mechanisms by which it works.

 

 16             Five years ago, at American Heart, Michael

 

 17   organized a session, and there were probably five

 

 18   or six people talking about this.  If you look at

 

 19   American Heart now, there are two days of people

 

 20   talking about this.  People didn't believe it five

 

 21   years ago.

 

 22             Now we have a critical mass in the field

 

 23   and we can start asking those questions, and I

 

 24   think the data will emerge over the next couple of

 

 25   years.

 

                                                               211

 

  1             DR. RAO:  Dr. Ruskin.

 

  2             DR. RUSKIN:  Doris, you described a very

 

  3   significant prolongation of action potential

 

  4   duration in the myoblasts from about 20 to 120

 

  5   milliseconds.  That suggests a change in iron

 

  6   channel expression.  Do you have any information as

 

  7   to how that came about?

 

  8             DR. TAYLOR:  Gus Grant told me it did?

 

  9   No.

 

 10             DR. RUSKIN:  I will buy that.

 

 11             DR. TAYLOR:  Short answer is no except

 

 12   that we know that we started seeing a plateau phase

 

 13   which wasn't there before, and we didn't change the

 

 14   rate of rise of the action potential.  Do I know

 

 15   any more than that?  No.

 

 16             Have we looked for channel markers?  You

 

 17   know, here I am saying, well, the only thing I will

 

 18   believe that if you tell me it's a cardiocyte, is a

 

 19   channel marker, have we looked for channel markers?

 

 20   No, because we did all of that in rabbits, and the

 

 21   darn markers don't exist.

 

 22             Are we now trying to figure that out in

 

 23   some of these other animal models?  Yes, and I

 

 24   think that is where some of the mice genetic models

 

 25   of changes in electrical activity in the heart may

 

                                                               212

 

  1   actually be really useful in terms of trying to

 

  2   dissect what myoblasts can do.

 

  3             DR. HARLAN:  You had a great quote from

 

  4   Einstein.  I will have to give you a quote I have

 

  5   from Osler, where he talks about stern iconoclastic

 

  6   spirit that we need, and that you reflected.

 

  7             My question is I think I misunderstood,

 

  8   you implied that there was not the cellular

 

  9   specificity that people assume, that you have seen

 

 10   some contractility with myoblasts, but also with

 

 11   bone marrow.  I wonder how extensively you have

 

 12   studied that with other cell types.

 

 13             DR. TAYLOR:  We have looked with

 

 14   myoblasts, we have looked with bone marrow stroma,

 

 15   and we have looked with bone marrow mononuclear

 

 16   cells, and we see an improvement with all of those.

 

 17   We haven't gone back and dissected the bone marrow

 

 18   mononuclear cell populations, we are starting to do

 

 19   that now.

 

 20             Other people have seen the same thing with

 

 21   MAPC cells.  So, have we dissected that in detail?

 

 22   No, but by the criteria that we have used, which is

 

 23   sonomicrometry and MRI, we see the effect, and yet

 

 24   we don't, when we look at histology, we don't see

 

 25   the same degree of muscle formation with all of

 

                                                               213

 

  1   those different cell types, which begins to argue

 

  2   that mechanism is more complicated than we thought.

 

  3             DR. HARLAN:  Let me ask it this way.  Are

 

  4   there cells that you have looked at, that you

 

  5   inject, where they don't work?

 

  6             DR. TAYLOR:  Fibroblasts.

 

  7             DR. HARLAN:  Fibroblasts don't work.

 

  8             DR. TAYLOR:  Fibroblasts actually improve

 

  9   compliance, but make systolic function worse in our

 

 10   hands.

 

 11             DR. RAO:  Dr. High.

 

 12             DR. HIGH:  I want to ask one question to

 

 13   try to reconcile some of your data preclinical

 

 14   studies with some earlier clinical work that we

 

 15   heard about.

 

 16             Was that in rabbits or pigs that you said

 

 17   that you needed at least 10                                              

                             8 cells to see an

 

 18   effect, was that rabbits?

 

 19             DR. TAYLOR:  That was rabbits--no, 3 x 10                      

                                                                               

            7

 

 20   in rabbits, 108 was in pigs, I am sorry.

 

 21             DR. HIGH:  Okay.  Then, the 10                                 

                                                                9 cells that

 

 22   are being injected in the clinical study would be

 

 23   roughly appropriately correlating in terms of--

 

 24             DR. TAYLOR:  Right, we actually see a

 

 25   better effect with 3 x 10                                                

                       8 cells in pig.  We

 

                                                               214

 

  1   haven't gone up to 10                                                    

            9 cells yet although we have

 

  2   plans to do that.  Philippe can tell you, you start

 

  3   dealing with massive numbers of cells, and when you

 

  4   are doing this in an autologous way, and you are

 

  5   dealing with cells that you have to keep at low

 

  6   confluence, it gets out of hand pretty quickly.

 

  7             DR. HIGH:  You said that in, is it the pig

 

  8   studies, about 15 to 20 percent of the injected

 

  9   cells are retained?

 

 10             DR. TAYLOR:  That is actually not data

 

 11   from my lab, that is data from other groups, and

 

 12   that has been in some pig studies, and I believe in

 

 13   some--I know of pig, and I can't remember what

 

 14   else.

 

 15             In our hands, in rabbit, we see a little

 

 16   bit higher than that, on the order of 20 to 25

 

 17   percent, but that has only been in a few studies

 

 18   with indium-labeled cells, so I don't trust those

 

 19   numbers yet.

 

 20             DR. HIGH:  Is that known to be a function

 

 21   of time after injury?

 

 22             DR. TAYLOR:  Actually, what we found is

 

 23   that the longer we wait after injury, the easier it

 

 24   is to get cells to hang around in the heart, that

 

 25   if we inject cells at 2 weeks, we get fewer cells

 

                                                               215

 

  1   retained than if we inject cells at a month.

 

  2             What we also know is if we inject cells in

 

  3   normal heart, they all go in the cardiac vein and

 

  4   get carried elsewhere, that the junctions in the

 

  5   myocardium are so tight that it is really hard to

 

  6   get those cells into the normal heart.

 

  7              Again, it gets back to injection.  We

 

  8   have come up with a way where we inject the cells,

 

  9   we see a bleb, we wait for the bleb to go away, we

 

 10   inject more cells, but that is just empiric,

 

 11   because it works for us.  Do we know how it is

 

 12   being done by other groups?  No clue.

 

 13             DR. SCHNEIDER:  Doris, you summarized

 

 14   nicely both the cellular complexities and the

 

 15   technical complexities that are involved here, and

 

 16   I wanted to comment about one in each of those

 

 17   categories.

 

 18             You talked about cryoinjury versus

 

 19   coronary artery ligation, and I wanted to agree

 

 20   with your comment that relatively little of the

 

 21   work in the field is being done with ischemia

 

 22   reperfusion injury, which more closely resembles

 

 23   the clinical situation particularly in an era of

 

 24   stenting and reperfusion therapies.

 

 25             A further complication there, though, is

 

                                                               216

 

  1   that much as was learned over a period of years in

 

  2   investigations of stunning, it may be necessary to

 

  3   distinguish between open-chested ischemia

 

  4   reperfusion injury and the chronically instrument

 

  5   close-chested animal that undergoes ischemia

 

  6   reperfusion injury, which is something that a few

 

  7   labs have been able to develop as a means to

 

  8   minimize potential artifactual effects of the

 

  9   surgical procedures.

 

 10             DR. TAYLOR:  Right.

 

 11             DR. SCHNEIDER:  With respect to cellular

 

 12   heterogeneity, you talked about the possibility

 

 13   that SP cells in the skeletal muscle population

 

 14   might be important. Michael Rednicke has identified

 

 15   in skeletal muscle, and investigators at Indiana

 

 16   have, as well, a scar-positive, LIN-negative,

 

 17   CD34-negative, CD45-negative population that lacks

 

 18   any ability to undergo hematopoietic

 

 19   differentiation and very closely resembles the

 

 20   scar-positive cells we found in adult heart.

 

 21             DR. TAYLOR:  I have seen data from the

 

 22   University of Minnesota like that, as well, and I

 

 23   also should say that Johnny Heward at Pittsburgh

 

 24   has found that as you increase the passage number

 

 25   of cells, and these are actually old data, from '98

 

                                                               217

 

  1   I believe, that as you increase the passage number

 

  2   of cells in vitro that you see differences, I think

 

  3   passages 3 and passage 5, or something like that,

 

  4   give you much better functional results in

 

  5   engraftment than passages 1, 2, and 4, and the

 

  6   desmin staining of those changes.

 

  7             So, I think we probably are selecting for

 

  8   different cells.

 

  9             DR. SCHNEIDER:  Along those lines, do you

 

 10   know if the scar-1-positive fraction goes up or

 

 11   goes down in your skeletal muscle cells over time?

 

 12             DR. TAYLOR:  Scar-1, we haven't look at

 

 13   scar-1 in our population of cells.  We have looked

 

 14   primarily for SP cells.  What I will say is that we

 

 15   grow our cells a little differently than most

 

 16   people.  As you know, we make an explant and

 

 17   actually allow our cells to grow out from the

 

 18   explant, and we are  getting a much higher

 

 19   percentage of more immature cells than other people

 

 20   as a result, and I think that is because we are not

 

 21   throwing them all away when we do the filtration

 

 22   and enzyme digestion, and those kind of things, and

 

 23   that may impact some of our functional data.

 

 24             DR. RAO:  We will take on last question.

 

 25   Dr. Simons.

 

                                                               218

 

  1             DR. SIMONS:  Doris, you mentioned that up

 

  2   to 90 percent of cells that they injected in the

 

  3   heart die soon thereafter.  So, the cells that are

 

  4   still there, do they need to be concentrated at a

 

  5   certain per sort of square area of the cell, or can

 

  6   you spread them as much as you like, so is it

 

  7   really a mechanical effect or is it something that

 

  8   the cells make, because you are making a point that

 

  9   it matters where you actually inject them?

 

 10             DR. TAYLOR:  What we have found is that we

 

 11   can do three parallel injections, and we get the

 

 12   same effect as if we do one injection, as if we do

 

 13   a star-shaped injection, so we spread them out in

 

 14   different--I mean we have given them under

 

 15   different geometries, and we see the same effect,

 

 16   the same dose of cells.

 

 17             What we don't know is how many die under

 

 18   each of those conditions.

 

 19             DR. SIMONS:  So, if you can spread them

 

 20   around, that would imply that that this is probably

 

 21   not a purely mechanical effect?

 

 22             DR. TAYLOR:  I think there is absolutely

 

 23   some truth to that.  I don't think it's purely

 

 24   mechanical.  I also don't think--that is what I

 

 25   said when we use these muscle cells, there are

 

                                                               219

 

  1   multiple populations of cells in there, and I think

 

  2   different ones have different effects.

 

  3             Absolutely, I think mechanism is an open

 

  4   question, we really don't know.

 

  5             DR. RAO:  In the interests of time, I

 

  6   guess we move on.   Thank you, Doris.

 

  7             I apologize for not recognizing members of

 

  8   the public, but this is the part of the meeting

 

  9   where we have to give priority to the committee

 

 10   members in terms of questions.  People in the

 

 11   audience can address the committee in the open

 

 12   session.

 

 13             Dr. Itescu.

 

 14                Preclinical Models - Hematopoietic and

 

 15         Mesenchymal Cell Therapies for Cardiac Diseases

 

 16             DR. ITESCU:  Thank you very much for

 

 17   inviting me here today.  As if the talks haven't

 

 18   been complex enough, I have got the difficult task

 

 19   of speaking for 30 minutes and covering a variety

 

 20   of animal models, as well as a variety of cell

 

 21   types, so I hope it will be cohesive enough.

 

 22              The issues to consider in this field, I

 

 23   have tried to address some of them here in this

 

 24   slide in terms of small animal models or large

 

 25   animal models, and it is the precise

 

                                                               220

 

  1   characterization of cell type and population to be

 

  2   used to define the cell source and process for

 

  3   isolation to determine if there is a need for ex

 

  4   vivo culture and expansion to identify the

 

  5   mechanisms of action for inducing cardiac repair,

 

  6   to identify appropriate animal models, in other

 

  7   words, small versus large, species-specific versus

 

  8   those that use human products and those that

 

  9   involve acute versus chronic ischemia.  These are

 

 10   all very, very important  questions.

 

 11             Finally, experiments that address

 

 12   dose-ranging studies for functional correlation and

 

 13   toxicity, and the last question which has not been

 

 14   touched on yet today, which I will at the end of my

 

 15   talk, is that between autologous versus allogeneic

 

 16   products.

 

 17             The adult mammalian bone marrow contains

 

 18   two stem cell populations at least.  The one that

 

 19   we are most familiar with are hematopoietic stem

 

 20   cells defined as being CD34-positive, and more

 

 21   importantly, CD45-positive.

 

 22             These cells form blood-forming elements,

 

 23   such as monocyte and macrophage lineage cells,

 

 24   these account for about 10 to 20 percent of the

 

 25   CD45-positive fraction, and more recently, cells

 

                                                               221

 

  1   that are endothelial progenitor cells or

 

  2   angioblasts that express these markers plus several

 

  3   others such as AC133 and c-kit.

 

  4             The second population, the

 

  5   non-hematopoietic stem cell fraction is typically

 

  6   CD45-negative and CD34-negative, and at least three

 

  7   cell types have been defined within this fraction -

 

  8   the mesenchymal stem cells, which really is poorly

 

  9   termed as stem cells since these cells are defined

 

 10   based on their in-vitro culture characteristics.

 

 11   The way they are isolated is very crudely, very

 

 12   grossly based on density properties and plastic

 

 13   adherence.  In fact, the population cells that is

 

 14   pulled out initially is very heterogeneous.

 

 15             A second stem cell type is the MAPC that

 

 16   you have heard about.  These cells are potentially

 

 17   more homogeneous in nature, however, the fact that

 

 18   they are dependent on negative immunoselection

 

 19   means that again we don't really know what the cell

 

 20   type that is ultimately derived is, and the cell

 

 21   culture conditions are very laborious and require

 

 22   low density for outgrowth.

 

 23             Finally, there is a third population of

 

 24   mesenchymal precursor cell or progenitor cell which

 

 25   can be defined on the basis of several surface

 

                                                               222

 

  1   markers and can be selected by immunoselection,

 

  2   positive immunoselection freshly from bone marrow,

 

  3   and we are using these cells currently in my

 

  4   laboratory, and I will touch on them a little bit

 

  5   towards the end of the talk.

 

  6             In respect to the hematopoietic stem cell

 

  7   fraction, the CD34/CD45 fraction, there is high

 

  8   frequency within the bone marrow compartment, as I

 

  9   have mentioned, 10 to 20 percent of macrophages, 1

 

 10   to 2 percent of the CD34 progenitor cells that can

 

 11   be freshly isolated without any requirement for

 

 12   in-vitro culture and expansion, and are well

 

 13   characterized for many years with established

 

 14   isolation protocols.

 

 15             In this slide, I tried to summarize the

 

 16   interaction and cooperation between the

 

 17   CD45-positive and 45-negative subsets in terms of

 

 18   vascular network formation, so that in the bone

 

 19   marrow, the primordial circle stem cell may give

 

 20   rise to the CD45-positive/34-positive endothelial

 

 21   progenitor cells, which egress to the embryonic

 

 22   organs where you get the initial vasculogenic

 

 23   small, thin-walled capillary development.

 

 24             At the same time, the CD45-negative

 

 25   parasite fraction derived from a mesenchymal

 

                                                               223

 

  1   progenitor cell produces a variety of factors

 

  2   including VEGF, FGF, angiopoietin, and SDF-1, which

 

  3   can interact with these vasculogenic capillaries to

 

  4   give rise to the more mature vascular network

 

  5   through a process of angiogenesis.

 

  6             In addition, the parasites may also

 

  7   migrate and give rise to the smooth muscle outer

 

  8   wall, so that you really have this cooperation

 

  9   between both cells and factors to give rise to the

 

 10   permanent new vessel formation.

 

 11              With this as a background, I will like to

 

 12   address several studies that have looked at how

 

 13   angiogenesis per se can improve cardiac function.

 

 14   This study from Kobashi [ph] and Collins in 2000, I

 

 15   think was the first to demonstrate or one of the

 

 16   first to demonstrate that whole bone marrow in a

 

 17   small rodent model could induce transient

 

 18   angiogenesis and improve cardiac function.

 

 19             You can see here the implantation of whole

 

 20   bone marrow into the ischemic myocardium of a rat

 

 21   gives an increase in local production of VEGF

 

 22   protein within 24 hours, but by 7 days this protein

 

 23   production is gone, and in parallel, the

 

 24   angiogenesis that is occurring in the heart is

 

 25   again transient, maximal at 1 week

 

                                                               224

 

  1   post-implantation, gone by 4 weeks.

 

  2             So, I think this particular model gives us

 

  3   some pause and suggests that the whole bone marrow

 

  4   approach may not be a way to give rise to permanent

 

  5   vasculature.  Others have used this sort of

 

  6   approach now in larger models, in pigs.  Work from

 

  7   Kemiharder [ph] and colleagues demonstrated that in

 

  8   a LAD-ligated porcine model, again whole bone

 

  9   marrow implantation gives rise to angiogenesis.

 

 10   This is within 6 weeks post-implantation, you can

 

 11   see improvement in collateral flow associated with

 

 12   some degree of improvement in function.

 

 13             In parallel studies they published about

 

 14   two years ago now, the same group demonstrated that

 

 15   in a more chronic ischemia model in pigs using the

 

 16   ameroid constrictors, they were able to again

 

 17   induce angiogenesis using mononuclear cells from

 

 18   bone marrow, as well as mononuclear cells from

 

 19   peripheral blood to a lesser extent.  You can see

 

 20   improvement in regional blood flow and improvement

 

 21   in global parameters of cardiac function including

 

 22   ejection fraction DPDT, and a reduction in the

 

 23   ischemic area.

 

 24             But again, all of these studies were done

 

 25   in the setting of an acute ischemic or perhaps more

 

                                                               225

 

  1   subacute ischemic model in this scenario, and the

 

  2   animals were followed up for no more than 6 weeks,

 

  3   so if we take that into consideration that in the

 

  4   smaller rodent, the neovascularization and the

 

  5   cardiac function improvement was transient, but I

 

  6   think the 6-week follow-up in these larger models

 

  7   is really not adequate especially in a pig where

 

  8   the physiology much more closely resembles that in

 

  9   man.

 

 10             The next approach is to look at perhaps

 

 11   subsets of some of these cells, and I would like to

 

 12   show you some work on endothelial progenitor cells

 

 13   defined by surface markers.

 

 14             Isner and colleagues originally described

 

 15   these cells demonstrating that endothelial

 

 16   progenitor cells were present in the bone marrow,

 

 17   were released to the circulation under certain

 

 18   signaling and present from growth factors under

 

 19   ischemic conditions, and that these cells can

 

 20   incorporate into regions of ischemia.

 

 21             So, we asked the question if one could

 

 22   identify these cells in the adult marrow of humans,

 

 23   could they potentially improve function through a

 

 24   process of neovascularization.

 

 25             The model that we use in my lab is one of

 

                                                               226

 

  1   using the incompetent nude rat, which is an athymic

 

  2   rat model that lacks T cells and B cells, continues

 

  3   to have some degree of natural killer cell

 

  4   function, in other words, it is a linking model,

 

  5   but it is able to tolerate certain types of human

 

  6   cell injections, particularly human cells that have

 

  7   not been in vitro cultured and expanded.

 

  8             Our objective was to cause a permanent

 

  9   occlusion of the anterior descending left coronary

 

 10   artery, in contrast again to other models that

 

 11   perhaps are using the reperfusion type of model.

 

 12   Our objective was to induce an infarct and see

 

 13   whether we could then protect the subsequent

 

 14   territories of myocardium still at risk in the

 

 15   periphery.

 

 16             We mobilized in the progenitor cells from

 

 17   healthy human donors using GCSF, think that, in

 

 18   fact, in the future  this would have been a way to

 

 19   move towards clinical trials, in other words, being

 

 20   able to harvest large numbers of progenitor cells.

 

 21             In our studies, we then immunoselected

 

 22   these on the basis of surface markers, CD34 and

 

 23   CD117, injected these into the tail vein of acute

 

 24   ischemic rats to see whether they homed to the

 

 25   myocardium.

 

                                                               227

 

  1             I would just like to pause for a minute to

 

  2   say that given the recent paper in the Lancet, I

 

  3   think a week or two weeks ago, I think that

 

  4   certainly raises a question about how such cells

 

  5   are going to be accessed in large numbers if they

 

  6   are to be used at all at the time of an acute

 

  7   myocardial infarct because I think under a local

 

  8   anesthetic, a bone marrow aspirate gives rise to

 

  9   extremely few numbers of these types of cells, and

 

 10   you really do require either large harvesting or

 

 11   large numbers to be mobilized, and I think at this

 

 12   point in time, GCSF is not the agent to be used

 

 13   post-infarct.

 

 14             In any case, the point of our studies were

 

 15   to see whether intravenous administration at the

 

 16   time of acute infarct would lead to selective

 

 17   homing to the myocardium, and we have met these

 

 18   trafficking pathways based on the type of chemokine

 

 19   expression that occurs post-infarct, and we found

 

 20   that B cells selectively migrate to the

 

 21   peri-infarct region, where they within two weeks

 

 22   induce both incorporation into vessels, and this is

 

 23   staining with antihuman CD31, and you can see cells

 

 24   that are previously labeled with dye/Dil,

 

 25   incorporate into vessels of the peri-infarct

 

                                                               228

 

  1   region, but in addition, a very dense induction of

 

  2   angiogenesis at the more distal areas as defined by

 

  3   rat-specific anti-CD31 antibodies.

 

  4             So, again, akin to the type of data shown

 

  5   by Dr. Epstein, it appears that these cells are

 

  6   able to not only incorporate and induce

 

  7   vasculogenesis, but presumably secrete a variety of

 

  8   antigenic factors.

 

  9             From a pathophysiologic perspective, we

 

 10   see that when we induce a vascular network at the

 

 11   peri-infarct region, you can see these large

 

 12   capillaries.  There is nice viability of the

 

 13   cardiomyocytes at the peri-infarct region as

 

 14   opposed to these apoptotic cardiomyocytes.  You see

 

 15   reduction in scar formation and clearly a viability

 

 16   and survival of the myocytes.

 

 17             But in addition to that, we were quite

 

 18   surprised to see that as early as five days

 

 19   post-neovascularization, there seems to be

 

 20   induction of cell cycling by endogenous

 

 21   myocyte-like cells, and you can see these by

 

 22   confocal microscopy small cells of the peri-infarct

 

 23   region that express alpha-sarcomeric actin in blue,

 

 24   but more interestingly, in yellow, the staining

 

 25   with the rat-specific anti-KR67 antibody, which

 

                                                               229

 

  1   recognizes cycling cells only of rat origin.  You

 

  2   can see lots of these type of small cells in the

 

  3   peri-infarct region only in the situation where we

 

  4   also have neovascularization being induced by the

 

  5   human cells.

 

  6             Within two weeks, in these same areas,

 

  7   what you see is that the cells that express

 

  8   sarcomeric actin now express troponin, and you can

 

  9   see an example of that large mature differentiated

 

 10   cardiomyocyte where the nucleus continues to be in

 

 11   cell cycle, and we see about 8-fold increase in

 

 12   numbers of these type of cells at the peri-infarct

 

 13   region that has received the human progenitor

 

 14   cells.

 

 15             We think that the cells or the new

 

 16   capillaries are secreting factors that are

 

 17   presumably inducing cycling of these

 

 18   cardiomyocytes, and we are looking at a variety of

 

 19   anti-apoptotic genes that are increased in

 

 20   expression in these cells including redox-related

 

 21   anti-apoptotic genes.

 

 22             The end result is a very significant

 

 23   salvage of the anterior wall of myocardium.  You

 

 24   can see that here, and you can see in this scenario

 

 25   obviously a very dramatic left ventricular scar

 

                                                               230

 

  1   formation and aneurysmal formation here.

 

  2             We can show a dose-dependent reduction in

 

  3   scar formation at the left ventricle as we increase

 

  4   the number of progenitor cells that we inject.

 

  5             Now, others have published recently that

 

  6   perhaps the CD34 cells may have the ability to

 

  7   become multipotential, perhaps transdifferentiate,

 

  8   however, the data are fairly scant, and I would say

 

  9   that given that only 1 in 7 healthy animals

 

 10   demonstrated HLA human troponin co-staining, I

 

 11   think at this point, it is open to debate whether,

 

 12   in fact, these cells do transdifferentiate or

 

 13   whether these cells have the capability to simply

 

 14   fuse with the donor cells.

 

 15             Nevertheless, whatever the precise

 

 16   mechanism, it appears that hematopoietic stem cell

 

 17   injection, either intravenously or

 

 18   intramyocardially, does result in significant

 

 19   global improvement in cardiac function, as we

 

 20   showed here, at least 30 percent improvement in

 

 21   ejection fraction, which is permanent.  This was 15

 

 22   weeks of follow-up post-LAD ligation, and we can

 

 23   look at fractional shortening or DPDT and

 

 24   demonstrate similar sort of things.

 

 25             In this scenario, you can see that there

 

                                                               231

 

  1   are other cell types that we use as controls,

 

  2   CD34-negative cells, saphenous vein endothelial

 

  3   cells, and what this sort of study shows is that

 

  4   you must always use different cell types when you

 

  5   are trying to evaluate a particular therapy.  It

 

  6   cannot just be compared to saline or existing

 

  7   control.  You have got to demonstrate specificity

 

  8   in the product that is being tested.

 

  9              Moreover, the question that we asked was

 

 10   what about if we combine this type of an approach

 

 11   with existing therapies, because really that is the

 

 12   question you want to address, and the example of

 

 13   restenosis with stenting is just one type of

 

 14   combination therapy between a new product and

 

 15   existing therapies.

 

 16             What you see here is if we combined CD34

 

 17   cells with both ACE inhibitors and beta blockers in

 

 18   the same rodent model, we had very significant

 

 19   synergy in outcome, and histologically, the reason

 

 20   for that was really not because the two products

 

 21   worked in a similar way, but because they had very

 

 22   separate mechanisms.

 

 23             The ACE inhibitors and beta blockers had

 

 24   no impact on neovascularization, but, in fact,

 

 25   prevented fibrosis in the posterior wall as they

 

                                                               232

 

  1   are known to do, so we have two very different

 

  2   mechanisms of action working in synergy, but it

 

  3   could have just as easily worked the other way

 

  4   around, and that is why we did the experiment.

 

  5             It could have been that by reducing wall

 

  6   strain or reducing fibrous replacement, we may have

 

  7   reduced the drive for endothelial cells to induce

 

  8   neovascularization and improve cardiac function.

 

  9             So, what can be concluded from preclinical

 

 10   data  using non-cultured cells?  The objectives

 

 11   that one should look for when you do these kind of

 

 12   studies include identifying the predominant

 

 13   mechanism by which a given cell type induces

 

 14   cardiac recovery.

 

 15             A comparison of efficacy of one given cell

 

 16   type with others, identification of the tissue

 

 17   distribution of that cell type following the

 

 18   preferred mode of delivery, unique short- or

 

 19   long-term risks associated with the preferred cell

 

 20   type, unique risks associated with the method of

 

 21   cell acquisition or isolation.  I have given you

 

 22   the example of GCSF administration, and perhaps

 

 23   alterations in efficacy or safety following

 

 24   coadministration with standard therapies.

 

 25             Now, I would like to shift attention a

 

                                                               233

 

  1   little bit to the second population I want to talk

 

  2   about, the non-hematopoietic stem cells, the

 

  3   CD34-negative, CD45-negative fraction.

 

  4             These cells are rare.  They account for

 

  5   less than 0.01 percent of bone marrow cells, and

 

  6   they do require, for this reason, in-vitro culture

 

  7   and expansion.

 

  8             I apologize about the complexity of this

 

  9   slide.  I will just take you through it a little

 

 10   bit.  I mentioned these three types of mesenchymal

 

 11   lineage cells that people are working with.  The

 

 12   so-called mesenchymal stem cells are more likely

 

 13   committed progenitors, but the point is that they

 

 14   are tolemerase-negative.

 

 15             The multipotent adult progenitor cells are

 

 16   MAPCs, tolemerase-positive, and the stromal or

 

 17   mesenchymal precursor cells are also

 

 18   tolemerase-positive.  These cells may be related to

 

 19   each other, but all three types of cells give rise

 

 20   to mesoderm lineage cells.  The MAPCs have also

 

 21   been shown to give rise to endodermal and

 

 22   ectodermal cells. It appears that the mesenchymal

 

 23   precursor cells can also give rise to endoderm and

 

 24   ectoderm.

 

 25              With respect to mesoderm, which is really

 

                                                               234

 

  1   what we are interested in primarily here, cardiac

 

  2   muscle and smooth muscle lineage cells can both be

 

  3   differentiated from the mesoderm, but also these

 

  4   cells can give rise to other cell types with

 

  5   mesodermal lineage.

 

  6             The so-called mesenchymal stem cells are

 

  7   cells that are derived from whole bone marrow and

 

  8   then isolated by simple density centrifugation to a

 

  9   particular layer.  These cells are then taken from

 

 10   the interface, put down on plastic, adhered for 24

 

 11   to 48 hours, and then the cells that continue to

 

 12   adhere are then cultured again fairly crudely with

 

 13   generally fetal bovine serum for weeks at a time.

 

 14             So, if you understand how this process is

 

 15   initiated, you understand that really, the

 

 16   isolation of these cells is so crude that you are

 

 17   starting out with a very heterogeneous population

 

 18   simply based on density characteristics, and that

 

 19   the true multipotential cell within this fraction

 

 20   probably is not more than 1 in 1,000 to 1 in 10,000

 

 21   of the cells you are starting out with.

 

 22             In any case, after, say, 10 to 14 days of

 

 23   culture and passage, you see the sort of

 

 24   monomorphous type of fibroblastoid phenotype, and

 

 25   you see the cells that have survived this period of

 

                                                               235

 

  1   culture are fairly homogeneous in terms of the type

 

  2   of markers that are being used.

 

  3             There are specific antibodies that can

 

  4   define surface characteristics of these cells, SH2,

 

  5   SH3, endoglin, and I am not sure what SH3 actually

 

  6   defines.  But in any case, these cells, after two

 

  7   weeks in culture, become fairly homogeneous.

 

  8             I borrowed this slide from Dr. Epstein,

 

  9   his recent paper in Circulation Research, which

 

 10   shows that these cells, after several passages in

 

 11   culture, demonstrate production following induction

 

 12   of ischemia of a variety of factors that are

 

 13   associated with both angiogenesis and

 

 14   arteriogenesis, and in a nice model or rat hind leg

 

 15   ischemia, you can see--I think you showed this

 

 16   slide already--demonstration of improvement in

 

 17   perfusion following MSC infusion in these cells in

 

 18   ligation of the hind leg artery.

 

 19             But in addition to secretion of

 

 20   pro-arteriogenic factors, the interest in these

 

 21   cells lies in the fact that they seem to have

 

 22   multipotential capability in that under appropriate

 

 23   culture differentiation conditions, they can be

 

 24   differentiated to adipocytes, chondrocytes, and

 

 25   osteocytes.

 

                                                               236

 

  1             Work now since at least 1999 has

 

  2   demonstrated that if you use appropriate inductive

 

  3   signals in these cells, in this case, 5-azacytidine

 

  4   to demethylate the cells, but there have been

 

  5   reports of other agents that can do similar sort of

 

  6   things, you can push the cells toward a

 

  7   cardiomyocyte-like lineage with appropriate marker

 

  8   expression and electromicroscopic criteria

 

  9   consistent with BT myotubes.

 

 10             Probably the best study to date to

 

 11   demonstrate that these cells can actually do

 

 12   something in vivo is work from Victor Dzaus' lab

 

 13   published last year in Nature Medicine.  Here, we

 

 14   are talking about again rat mesenchymal lineage

 

 15   cells cultured in the way I have just defined with

 

 16   density separation and long-term culture with

 

 17   bovine serum, fetal calf serum, and what you see is

 

 18   after injection of these cells into the

 

 19   peri-infarct region of rats following LAD ligation,

 

 20   these cells appear at least phenotypically to

 

 21   acquire markers of cardiomyocytes, so that they

 

 22   express myosin heavy chain, cardiac troponin,

 

 23   sarcomeric actin, and connexin 43.  This is within

 

 24   two weeks of implantation.

 

 25             However, the majority of these cells just

 

                                                               237

 

  1   don't survive when you put them in.  They die after

 

  2   early transplantation.  Causes appear to be

 

  3   ischemia again, competition for oxygen nutrients,

 

  4   inflammatory and oxidative stress in the

 

  5   post-infarct myocardium, loss of survival signals

 

  6   from cell to cell or cell matrix interactions, and

 

  7   probably lack of tolemerase activity and

 

  8   self-renewal capability by these cells, and I said

 

  9   to you earlier that these cells just don't express

 

 10   tolemerase, certainly when they are in culture.

 

 11             The work by Mongi, et al., in fact, showed

 

 12   that if you genetically modify these cells with an

 

 13   AKT transgene, you could significantly prevent

 

 14   these cells from dying following implantation,

 

 15   significantly reduce the apoptotic index, increase

 

 16   survivability, and overall improve function almost

 

 17   to the level of the non-infarcted animals, quite

 

 18   impressive, but it required essentially making them

 

 19   resistant to death or apoptosis by AKT

 

 20   overexpression.

 

 21             I will just move along to the second

 

 22   population of cells, the MAPCs.  These cells, as I

 

 23   mentioned, are defined based on negative expression

 

 24   of all known markers, CD45 and many other known

 

 25   markers of lineage-specific differentiation, and

 

                                                               238

 

  1   following long-term culture, these cells have been

 

  2   shown to be capable of differentiating to a variety

 

  3   of tissue types requiring cocktails of various

 

  4   cytokines and differentiation factors.

 

  5             In particular, following activation with

 

  6   VEGF and implantation into appropriate medium, in

 

  7   this case, tumor model in the scid mouse, these

 

  8   cells appear to be able to incorporate into first

 

  9   neovascularization, and you can see they can induce

 

 10   networks of capillaries, neocapillaries really, in

 

 11   wound-healing tissues, so they may contribute to

 

 12   vascularity.

 

 13             With that as a backdrop, Gallegos and

 

 14   colleagues at the University of Minnesota decided

 

 15   to look at whether again fresh non-differentiated

 

 16   MAPCs from dogs could be implanted into an acutely

 

 17   LAD-ligated model and could perhaps improve

 

 18   function in some form.

 

 19             The model was to induce again a complete

 

 20   LAD ligation in the dog, autologous cells were

 

 21   taken from the marrow, they were expanded for about

 

 22   four weeks, and then four weeks later, injections

 

 23   were put directly into the myocardium.

 

 24             What they found was that a month

 

 25   post-injection of the cells,  there appeared to be

 

                                                               239

 

  1   increase in myocardial perfusion reserve defined as

 

  2   perfusion under stress relative to perfusion at

 

  3   rest compared to the controls, and this is in four

 

  4   animals.

 

  5             But significantly, although there was some

 

  6   improvement in regional myocardial contractility

 

  7   within the infarct area and the peri-infarct zone,

 

  8   defined as circumferential shortening and radial

 

  9   thickening, there was actually no improvement in

 

 10   global function of cardiac measurements as defined

 

 11   through systolic improvement by fractional

 

 12   shortening or ejection fraction.

 

 13             Now, just to the last area that I would

 

 14   like to touch on, and that is the possible use of

 

 15   cultured allogeneic stem cells for cardiac disease.

 

 16   The issues to consider here are whether or not

 

 17   cells constituitively or under inductive conditions

 

 18   express surface molecules, surface HLA molecules,

 

 19   and co-stimulatory molecules, secondly, whether

 

 20   stimulation of recipient T cell responses occur in

 

 21   vitro, and, thirdly, whether these cells might

 

 22   induce inflammatory responses after in vivo

 

 23   injection.

 

 24             This work may or may not be familiar to

 

 25   many people here, but many labs now have completely

 

                                                               240

 

  1   demonstrated that mesenchymal lineage cells from

 

  2   humans, from primates, and from smaller animals

 

  3   routinely actually do not express Class II HLA

 

  4   molecules until they are induced by

 

  5   gamma-interferon.  They certainly do all express

 

  6   Class I, but in addition to that, do not express a

 

  7   variety of co-stimulatory molecules, such as CD40,

 

  8   CD80, and CD86.

 

  9             All of these molecules are absolutely

 

 10   critical in inducing T cell immune responses when

 

 11   you transplant cell or an organ between

 

 12   individuals.

 

 13             What I want to show you here is that

 

 14   mesenchymal stem cells clearly do not induce a T

 

 15   cell response following standard mixed leukocyte

 

 16   reactions in vitro.  This, you can see in

 

 17   comparison to allogeneic mononuclear cells, which

 

 18   induce a vigorous T cell response.

 

 19             In addition, even activation with

 

 20   interferon gamma to upregulate Class II HLA has no

 

 21   effect on mesenchymal stem cell induction of T cell

 

 22   responses, where you can see the potency of

 

 23   gamma-interferon at inducing allogeneic responses

 

 24   if you do it to dendritic cells or to allogeneic

 

 25   mononuclear cells.

 

                                                               241

 

  1             So, there is something very special about

 

  2   these cells, that they do not seem to express

 

  3   co-stimulatory molecules, they do not seem to

 

  4   induce an allogeneic T cell response at least in

 

  5   vitro, and moreover, they seem to have the ability

 

  6   to suppress ongoing immune responses.

 

  7             What you can see here is that this is now

 

  8   a third party mixed leukocyte reaction where you

 

  9   have T cells from one donor proliferating.  This is

 

 10   to allogeneic mononuclear cells, to allogeneic

 

 11   dendritic cells, or to PHA, and you can see that if

 

 12   you add third party mesenchymal stem cells, they

 

 13   will suppress this proliferative response, and this

 

 14   suppressive effect is in a dose-dependent manner.

 

 15             Additional studies have suggested that, in

 

 16   fact, part of this suppressive effect requires

 

 17   cell-cell contact inhibition, and part of it

 

 18   requires secretion of a variety of

 

 19   anti-inflammatory cytokines, such as TGF-beta, but

 

 20   the precise mechanism at this point has not been

 

 21   defined.

 

 22             The only human trial that I am aware of

 

 23   using allogeneic mesenchymal stem cells to date is

 

 24   one where allogeneic stem cell transplants were

 

 25   performed with third party allogeneic mesenchymal

 

                                                               242

 

  1   stem cells, and I understand the results of those

 

  2   were better engraftment and reduction in

 

  3   graft-versus-host disease, suggesting again that

 

  4   the third party mesenchymal stem cell, not only

 

  5   does not induce an immune response, and is

 

  6   presumably not itself rejected, but is also

 

  7   beneficial in outcome.

 

  8             But just to go back to the cardiac studies

 

  9   now, using that again as the backdrop, Brett Martin

 

 10   and colleagues from Ceros Therapeutics--and these

 

 11   are two slices that he has given me--presented at

 

 12   the American Heart Association last year, their

 

 13   findings using allogeneic mesenchymal stem cells

 

 14   from pigs in acute myocardial injury, and this is

 

 15   now in a reperfusion model to persecute myocardial

 

 16   infarction, what they was generate a panel of

 

 17   allogeneic porcine mesenchymal stem cells that are

 

 18   matched at blood group antigen, they used very

 

 19   large numbers of these cells distributed over the

 

 20   infarct zone.  We are talking about a reperfusion

 

 21   model without any immunosuppression.

 

 22             What you see here is that within two weeks

 

 23   of implantation, the cells were still present, so

 

 24   they hadn't been rejected.  You can see the

 

 25   presence of cells two weeks later.  However, these

 

                                                               243

 

  1   cells did not differentiate to cardiomyocytes, so

 

  2   they did not induce a rejection episode, but they

 

  3   also did not appear to really do much in terms of

 

  4   integration or differentiation.

 

  5             With respect to function, what you see is

 

  6   actually improvement again in diastolic parameters,

 

  7   as we have heard earlier, with increasing diastolic

 

  8   wall thickness at various time points including

 

  9   well beyond six weeks of study, and a reduction in

 

 10   N-diastolic pressures, however, again no

 

 11   improvement in systolic function, which is again

 

 12   consistent with perhaps alterations in matrix,

 

 13   alterations in ground substance, but no real effect

 

 14   on contractility or myocytes.

 

 15             This table, what you see is really a

 

 16   summary of the type of the cells that are being

 

 17   used of mesenchymal lineage.  The MAPCs, so far we

 

 18   have seen only modest efficacy in cardiac models in

 

 19   dogs in the absence of predifferentiation in vitro.

 

 20             Mesenchymal stem cells have shown good

 

 21   function in rats, but modest only in pigs, and

 

 22   mesenchymal precursor cells are currently being

 

 23   investigated.  We think they may be far more potent

 

 24   than either of these two because you are able to

 

 25   isolate them by surface characteristics when they

 

                                                               244

 

  1   are fresh.

 

  2             The final slide here is that I think many

 

  3   remaining hurdles is the message.  The appropriate

 

  4   clinical indication needs to be defined.  The more

 

  5   proof of principle animal studies, we have got to

 

  6   determine optimal doses for efficacy, careful

 

  7   registry of adverse events in our animals, let

 

  8   alone humans.

 

  9             We have got to optimize the ex vivo

 

 10   culture process, which I haven't even talked about.

 

 11   The autologous versus allogeneic question is

 

 12   critical because it is going to impact both on the

 

 13   process and on the cost of this whole therapy,

 

 14   determine the best route of administration and

 

 15   really how do you improve engraftments, we have

 

 16   heard all about that before.

 

 17             Thank you very much.

 

 18             [Applause.]

 

 19             DR. RAO:  Thank you, Dr. Itescu, for a

 

 20   really nice summary and trying to help keep us on

 

 21   time, as well.

 

 22             In the interests of trying to get us back

 

 23   on track, I am going to ask people to limit their

 

 24   questions to things which are directly relevant to

 

 25   the talk and if there are specific burning

 

                                                               245

 

  1   questions on this issue.

 

  2                               Q&A

 

  3             DR. CUNNINGHAM:  I just have one question.

 

  4   I was curious, in all your rat studies you

 

  5   published in Nature, did the rats die, and what did

 

  6   they die of, you know, the ones that didn't get to

 

  7   the endpoint of the experiment, were there any

 

  8   adverse events that you noticed?

 

  9             DR. ITESCU:  You mean unrelated to the

 

 10   initial surgery?  We have something like 30, 40

 

 11   percent death rate from the initial surgery, but

 

 12   pre-cellular implantation, but after cellular

 

 13   implantation is what you are asking me, we carried

 

 14   the animals out.  There was no real adverse events

 

 15   that I am aware of in these studies.

 

                                                               246

 

  1             DR. KURTZBERG:  You made a passing comment

 

  2   as you were speaking that you thought GCSF wouldn't

 

  3   be safe after an MI.  Can you expand on that?

 

  4             DR. ITESCU:  I think in the recently

 

  5   published study, just to summarize my take on that

 

  6   study, a randomized study where one group received

 

  7   GCSF subcutaneously to mobilize the endogenous

 

  8   population of marrow, another group received GCSF

 

  9   subcutaneously to mobilize and then had the

 

 10   mobilized cells harvested, not immunoselected, and

 

 11   I think the cells were then delivered by I believe

 

 12  intracoronary routes, whole unfractionated cells.

 

 13             The conclusion of the study was that the

 

 14   patients that received the cell therapy had a

 

 15   significant improvement in cardiac function,

 

 16   whereas, the group that received GCSF alone did

 

 17   not.

 

 18             However, in both populations, the study

 

                                                               247

 

  1   was cut short because of this complication, but I

 

  2   think if you pooled both populations, 7 out of 10

 

  3   patients developed significant restenosis at the

 

  4   site of the stent implantation.

 

  5             Now, these are bare stents.  This is the

 

  6   pre-repamycin days.  I think the anticipated

 

  7   restenosis rate would have been maybe 25 percent at

 

  8   most in that population. I think even with

 

  9   repamycin, you would expect to reduce the rate--if

 

 10   70 percent is right--diabetic patients actually

 

 11   have a restenosis rate with repamycin stents still

 

 12   of about 20 to 25 percent, 50 percent without

 

 13   repamycin, so these patients are more severe than

 

 14   diabetics are.

 

 15             I wouldn't anticipate that you would be

 

 16   able to lower that to below 35, 40 percent, and

 

 17   that is obviously totally unacceptable.

 

 18             Now, the question is why is it that GCSF

 

 19   was associated with this high restenosis rate, and

 

 20   I think we can all make speculations, but I think

 

 21   at least one possibility is that the cells that are

 

 22   mobilized, CXER-4 positive cells from the marrow,

 

 23   many cell types express CXER-4 including smooth

 

 24   muscle progenitor cells, and I think that is

 

 25   probably the simplest explanation, that there are

 

                                                               248

 

  1   smooth muscle progenitors that are mobilized, that

 

  2   migrate to the site of the stent, and I am not sure

 

  3   how one can get around that actually, but that is

 

  4   just a guess really.

 

  5             DR. TAYLOR:  That was great.  I have one

 

  6   quick question.   You didn't mention this except

 

  7   that in vitro, a lot of these cells can become a

 

  8   number of mesodermal cell types, and I think there

 

  9   are some data in rats that showed early on that if

 

 10   you inject these cells in the center of an infarct,

 

 11   they, in fact, at times become adipocytes or

 

 12   chondrocytes or osteoblasts, or something like

 

 13   that.

 

 14             I wondered if you would comment on what

 

 15   cells you think are likely to do that and whether

 

 16   all of them are.

 

 17             DR. ITESCU:  Obviously, that is the worst

 

 18   case theoretical question in this whole field.

 

 19   What we don't want is to develop bone in the middle

 

 20   of their hearts.

 

 21             I am not aware really of a lot of studies

 

 22   that have demonstrated that sort of abnormal

 

 23   differentiation. Certainly, adipocytic

 

 24   differentiation has occurred, I am not sure that I

 

 25   have seen bone differentiation.

 

                                                               249

 

  1             It really depends on how well defined the

 

  2   cell population is, I think, and to what extent the

 

  3   cells have been predifferentiated or to what extent

 

  4   they may be still very multipotential.  It is not

 

  5   clear to me whether you need to start with a cell

 

  6   that is very multipotential or that is fully

 

  7   differentiated, and perhaps the culturing process

 

  8   where you are pushing the cells, and again using

 

  9   fetal calf serum I think is an unfortunate--it is

 

 10   the only thing that as been done to date--but it is

 

 11   I think probably the worst way to be culturing

 

 12   cells, because you don't know what is in  your

 

 13   culture medium, you are pushing these cells to many

 

 14   different lineages.

 

 15             When we look at these cells

 

 16   following--very few studies have been published

 

 17   that have looked at this--but we have looked

 

 18   ourselves in this way, and you can see that after

 

 19   three or four weeks of culture, you can see cells

 

 20   that express markers of mature smooth muscle cells,

 

 21   of mature, some bone differentiation, some

 

 22   differentiation to cartilage.

 

 23             Whether or not that is relevant when you

 

 24   put the cells back in, whether there is going to be

 

 25   differential potential for outgrowth of one cell

 

                                                               250

 

  1   type over another, I think is really a totally open

 

  2   question, but I would push towards putting cells

 

  3   that are less differentiated in rather than more

 

  4   differentiated, because I think the less

 

  5   differentiated cells have got the ability to still

 

  6   proliferate, to be pushed towards the appropriate

 

  7   lineage under appropriate inductive signals that

 

  8   may still be present in the heart.

 

  9             There may be more differentiation towards

 

 10   maybe a cardiomyocyte lineage, but at least towards

 

 11   a smooth muscle lineage, so you might get some

 

 12   degree of arteriogenesis, as well as I think it is

 

 13   the more undifferentiated cells that are the ones

 

 14   that produce the very rich supply of arteriogenic

 

 15   factors.

 

 16             Just getting back to what I was saying,

 

 17   the very undifferentiated cells express markers

 

 18   really of parasites, so if you are thinking of

 

 19   parasite implantation, it is one way of maintaining

 

 20   viability of endothelial cells and integrity of the

 

 21   endothelium, and maybe it is a way of building the

 

 22   vascular network.

 

 23             DR. RAO:  This is the last question.

 

 24             DR. BLAZAR:  The issue of

 

 25   transdifferentiation in vivo is striking that you

 

                                                               251

 

  1   can get cells there and they just sit there, and I

 

  2   think this has been seen with a number of

 

  3   non-hematopoietic cell sources, MSCs being one.

 

  4             I guess the question is whether there are

 

  5   inhibitory signals that are present that prevent

 

  6   differentiation in vivo under certain conditions,

 

  7   and has anyone ever taken these cells back out of

 

  8   the heart and show that they can, in fact, be

 

  9   induced to differentiate in vitro?  Do we know

 

 10   anything about the inhibitory factors present at

 

 11   the site that are precluding differentiation?

 

 12             DR. ITESCU:   Those are obviously great

 

 13   questions, very important questions, and again I

 

 14   think it speaks to having a good understanding of

 

 15   the surface markers of these cells because if you

 

 16   know what they express, you can do the sort of

 

 17   experiments that you are suggesting.  You know what

 

 18   you are putting in, and then you can actually take

 

 19   them out again based on maybe immunoselection.

 

 20             We are actually trying to do those type of

 

 21   experiments to ask exactly those questions, but I

 

 22   think in many ways you are limited then to human

 

 23   cells, because many of the well-defined surface

 

 24   markers, there just aren't enough reagents.

 

 25   Perhaps mouse cells is the only other.  The mouse

 

                                                               252

 

  1   system is the one that is well enough developed and

 

  2   the human system, and other than that, we are

 

  3   really missing reagents where you can do those type

 

  4   of experiments.               DR. RAO:  Thank you,

 

  5   Doctor.

 

  6             DR. HARLAN:  Can I still ask a quick

 

  7   question?  You were careful to say so in your talk

 

  8   when you were talking about allogeneic cells, and

 

  9   you said at least in vitro, these allogeneic cells

 

 10   don't activate T cells, but I just wish to

 

 11   emphasize the point that in vivo, it is so much

 

 12   more complicated than that, and the presence or

 

 13   absence of B7, I thought Bruce maybe was going to

 

 14   speak to this, you didn't make the point, but I

 

 15   think it is important to emphasize that whether or

 

 16   not a cell in vitro stimulates a T cell response is

 

 17   really a very poor predictor of whether that will

 

 18   be rejected in vivo.

 

 19             DR. ITESCU:  Well, I am not sure I agree

 

 20   with that at all actually.

 

 21             DR. HARLAN:  Then, we will have more

 

 22   discussion.  I saw the pig data, that is

 

 23   interesting.

 

 24             DR. ITESCU:  I am actually a transplant

 

 25   immunologist.  This is what I do, I take care of

 

                                                               253

 

  1   cardiac transplant patients, and I am going to tell

 

  2   you that 12 months ago, when I first heard about

 

  3   these sort of data, I was extremely skeptical,

 

  4   along similar lines to what you are saying, you

 

  5   know, is it an in vitro artifact.

 

  6             However, you go back and you see how many

 

  7   labs are reproducing these data, which is really

 

  8   what I am surprised about, and I can tell you that

 

  9   the mixed leukocyte reaction is about the best

 

 10   single assay that we have in transplant medicine to

 

 11   predict allogeneic rejection and allogeneic

 

 12   sensitization.

 

 13             In the old days, it used to be used

 

 14   routinely for kidney transplant selection,

 

 15   donor-recipient selection, and it is still the best

 

 16   assay.  I am not sure whether it reflects whether

 

 17   these cells are going to be accepted long term in

 

 18   vivo, but it is certainly a very good marker for

 

 19   biology.  I don't know what it means, but it is

 

 20   routinely being reproduced using these type of

 

 21   cells.

 

 22             DR. HARLAN:  To say it is the best, and I

 

 23   won't disagree with it, is fine, but it is still a

 

 24   very poor predictor of in vivo function, and it

 

 25   goes back 30 years to the two-signal model,

 

                                                               254

 

  1   thinking if you could get rid of the antigen

 

  2   presenting cells within a graft, that you would

 

  3   take, and it seems to work in rodents, but it just

 

  4   doesn't work in  higher animal models.

 

  5             Bruce, do you want to step in?

 

  6             DR. BLAZAR:  I think there is the point of

 

  7   somewhere in between because clearly, there are

 

  8   cell-to-cell contact phenomenon that happen in

 

  9   vitro with regulatory T cells.  You can show a

 

 10   TT-dependent inhibition of responses throughout

 

 11   TGF-beta, and depending on the models in vivo,

 

 12   those either are true or they fall apart.

 

 13             We know in matched sibling donor

 

 14   transplants, mixed leukocyte reaction culture does

 

 15   not predict graft versus host disease.  So, I think

 

 16   that if you do see suppression, it is encouraging

 

 17   to try to go forward for in vivo, and that is

 

 18   probably as good as you are going to do, but it

 

 19   doesn't necessarily mean that those mechanisms take

 

 20   place in vivo particularly as those cells

 

 21   themselves change in their own ability to elaborate

 

 22   cytokines or express other molecules.

 

 23             DR. ITESCU:  I agree entirely and I am

 

 24   saying the exact same thing, that I think it is a

 

 25   phenomenon that is reproducible in vitro, it's an

 

                                                               255

 

  1   unusual and unique scenario. Whether or not it has

 

  2   implications in vivo remains, but I think at least

 

  3   it ought to be tested, and I think it may have

 

  4   implications on dendritic tolerance-inducing

 

  5   mechanism, which truly is a different way of

 

  6   thinking about this all together.

 

  7             It may have nothing to do with the ability

 

  8   of these cells themselves to escape surveillance in

 

  9   the periphery, but perhaps they end up in the

 

 10   thymus, and they may actually be able to reeducate

 

 11   the immune response, but I think all of that

 

 12   remains open to test.

 

 13             DR. RAO:  Last comment.

 

 14             DR. NEYLAN:  A very quick

 

 15   transplant-related question, again, maybe bring

 

 16   some of these last discussions to a very practical

 

 17   safety concern.

 

 18             That is, given the ability of these

 

 19   allogeneic cells to abrogate or reduce the immune

 

 20   response of the host, is it possible that the

 

 21   migration and homing of these cells may differ to

 

 22   autologous cells in a way, maybe akin to the

 

 23   micro-chimers and observations of solid organ

 

 24   transplantation, that potentially pose a safety

 

 25   risk to the use of allogeneic cells, homing to or

 

                                                               256

 

  1   disseminating and finding a welcome home in other

 

  2   tissues.

 

  3             DR. ITESCU:  I think that is a fair point,

 

  4   in other words, if they are immunoregulatory and

 

  5   you are injecting them and they find their way into

 

  6   the thymus, for example, will they induce a state

 

  7   of tolerance to an exogenous antigen that the

 

  8   patient sees at exactly the same time, which is the

 

  9   concern whenever we use an immunoregulatory new

 

 10   drug.

 

 11             Now, there are a couple of studies

 

 12   actually that have just recently been published

 

 13   that suggest that, in fact, while they induce

 

 14   tolerance to themselves and maybe tolerance to an

 

 15   alloimmune reaction, they don't seem to have

 

 16   induced tolerance to an exogenous pathogen, for

 

 17   example.

 

 18             Now, again, that is two studies, more work

 

 19   needs to be done, but I think the question is fair,

 

 20   it's an absolutely valid point, and obviously, you

 

 21   have to worry about that when you do your studies

 

 22   and follow up the patients very closely.

 

 23             DR. RAO:  We will visit it tomorrow.  We

 

 24   should move on.

 

 25             Dr. Taylor.

 

                                                               257

 

  1             From Mouse to Man:  Is it a Logical Step

 

  2                       for Cardiac Repair?

 

  3             DR. TAYLOR:  It is a logical step because

 

  4   I think it raises actually the issues that we are

 

  5   just talking about, whether or not data we get from

 

  6   rodents actually can be translated to humans or to

 

  7   larger animals.

 

  8             I also want to say actually, in terms of

 

  9   an apology, I realized when I sat down that one of

 

 10   the reasons my last talk was so disjointed was that

 

 11   the version that was up here was not the version

 

 12   that I had on my computer, so it was a kind of

 

 13   foreign talk to me, so I apologize and hope we will

 

 14   do better this time.

 

 15             From mouse to man, is it a logical step?

 

 16   I am going to start with another comment from

 

 17   Ghandi, which I have had on my office door for the

 

 18   last 10 years about this field, which is, "First,

 

 19   they ignore you, then, they laugh at you, then,

 

 20   they fight you, then you win."

 

 21              I think it raises the point that we are

 

 22   somewhere in the continuum in this field, and it is

 

 23   time for us to start asking the hard questions, so

 

 24   that we can have the fight and then win.

 

 25             As we are talking about moving from mouse

 

                                                               258

 

  1   to man, I think we have to talk about cell type,

 

  2   and I think I would be remiss if I didn't say that

 

  3   I think cell type depends on what we are trying to

 

  4   do here, and that if we are trying to look at

 

  5   chronic ischemia or hibernation, we are probably

 

  6   looking at cells that are more likely to induce

 

  7   angiogenesis, such as the cells you just heard

 

  8   about - bone marrow mononuclear cells, angioblasts,

 

  9   some subpopulation of stromal cells, growth

 

 10   factors, or maybe even myoblasts plus growth

 

 11   factors, but if you want functional repair and

 

 12   contractile cells, you are either want cells that

 

 13   are contractile, such as skeletal myoblasts or

 

 14   cardiocytes, maybe cardiac stem cells, or you want

 

 15   bone marrow cells that can become contractile

 

 16   cells.

 

 17             But the most important issue probably in

 

 18   this whole field, which is why I think we are going

 

 19   to have to talk about moving from mouse to man is

 

 20   probably this arrow, and the fact that we need both

 

 21   angiogenesis and myogenesis if we are going to have

 

 22   an appropriate outcome, and that, in fact, it may

 

 23   be not just one cell type, but multiple cell types

 

 24   that we end up needing for cardiac repair.

 

 25             Unfortunately, as we are looking at these

 

                                                               259

 

  1   cells, we don't have the opportunity to do those

 

  2   anywhere except in rodent at the present or in

 

  3   humans, so we are going to have to move from mouse

 

  4   to man at least with many of these cells unless

 

  5   industry provides us with the tools that we need to

 

  6   do the studies in between, because right now we

 

  7   really don't have the capability of moving to a

 

  8   larger animal model.

 

  9             I guess I want to start by asking the

 

 10   question what the appropriate preclinical animal

 

 11   models are and how quickly can we move forward by

 

 12   saying, you know, I presented this slide a minute

 

 13   ago, we had 15 years of preclinical data in rabbit

 

 14   and dog and pig and rat, mouse, and sheep, and we

 

 15   thought myoblast transplantation was safe,

 

 16   effective, and feasible.

 

 17             But we missed a lot of things, and we

 

 18   missed--I apologize, I thought there was another

 

 19   part down here, we are off to a great AV start, but

 

 20   that's okay--so what did we miss?  We missed the

 

 21   fact that these cells might be electrically

 

 22   incompatible with the remainder of the myocardium.

 

 23             We missed questions about location of

 

 24   injection, we missed questions about some of the

 

 25   dosing phenomena, we missed a number of things in

 

                                                               260

 

  1   our early preclinical models despite the fact that

 

  2   we used both large and small animal models.

 

  3             So, the question then really is what do

 

  4   you really need to do and when do you move to

 

  5   clinical studies, and I am going to give my jaded

 

  6   perspective for a minute and say that I think

 

  7   sometimes you move to clinical studies because your

 

  8   institution wants you to and kind of forces you to

 

  9   either because there is a financial incentive or

 

 10   that there is no such things as bad PR, but I would

 

 11   like to say that the appropriate time to move to

 

 12   clinical trials is when the data warrant it and

 

 13   that again we have to underpromise and overdeliver.

 

 14             So, if we believe that every cell injected

 

 15   seems to work, and that thus the heart is easier to

 

 16   repair than we thought, let's take, for example,

 

 17   the possibility that that is really the case, and

 

 18   if that is the case, when do we move to the clinic.

 

 19             I guess I could start by saying we have

 

 20   already moved to the clinic, but that being said,

 

 21   if we look at the clinical data, does it support

 

 22   the fact that the myocardium is easier to repair

 

 23   than we thought.

 

 24             Well, yes, everything works, but none of

 

 25   the clinical cell studies are placebo controlled,

 

                                                               261

 

  1   and 8 patients or even 53 patients can show you

 

  2   anything, especially when follow-up is short and we

 

  3   aren't considering age, gender, or heart failure

 

  4   status.

 

  5             I want to get back to these, and I will in

 

  6   this talk, that we haven't talked at all about

 

  7   factors like age and gender and how they may be

 

  8   really relevant, and there is a reason that drug

 

  9   trials involve thousands of patients with at leave

 

 10   five-year follow-up.

 

 11             Also, most of the clinical cell studies

 

 12   out there were designed as Phase I safety studies

 

 13   as they should have been, yet, many of these claim

 

 14   efficacy despite the fact that they were either

 

 15   revascularization studies or had other

 

 16   co-treatments involved, and I think it really

 

 17   raises questions about what we need to do.

 

 18             So, if we believe that the myocardium is

 

 19   easier to repair than we thought, what does that

 

 20   tell us about moving to clinical studies?  I am

 

 21   sorry, those two slides are actually backwards.  If

 

 22   every clinical cell works, what does that mean?

 

 23             I think it means that we have no clue how

 

 24   they work, whether they create angiogenesis or

 

 25   myogenesis, unloading of the heart, recruitment of

 

                                                               262

 

  1   stem cells or whatever.

 

  2             What questions does that raise?  It raises

 

  3   questions about patients, which raises the same

 

  4   questions about injury models, preclinical injury

 

  5   models.  It raises questions about dose and timing

 

  6   of cells, which then have to interact with the

 

  7   injury models.  It raises questions about route of

 

  8   administration or location of the cells, and how we

 

  9   measure the outcome, and those all affect which

 

 10   animal model you can choose.

 

 11             Well, the genie is out of the bottle, as

 

 12   the people at Mayo have said.  Clinical trials have

 

 13   started, so what do we do?  I think we educate

 

 14   people about what the appropriate situation is.

 

 15             There are a number of my clinical

 

 16   colleagues that I have talked with, and it scares

 

 17   me a little bit, who don't even realize that if you

 

 18   are going to use bone marrow derived cells, that

 

 19   the FDA needs to be involved if you are going to

 

 20   put them in the heart, and I think that is an issue

 

 21   that we really have to address.

 

 22              I think we need to require enough

 

 23   preclinical data, and then I think we need to quit

 

 24   rewarding people for doing it wrong.  What do I

 

 25   mean by that?  Well, what I mean by that is we have

 

                                                               263

 

  1   to quit doing science by the Washington Post, as

 

  2   you said earlier, and we have to quit focusing on

 

  3   the fact that this is a multibillion dollar market

 

  4   every year, and focus on the patients instead.

 

  5             So, what do myoblasts tell us about moving

 

  6   forward?  Well, as I said, what we knew and what we

 

  7   missed is that there are electrical events, the

 

  8   route of administration wasn't clear, location and

 

  9   timing wasn't clear, culture medium, we thought we

 

 10   knew, but it didn't turn out to always be the case.

 

 11             Autologous serum has been reported to be

 

 12   safer than non-autologous serum.  Different

 

 13   vehicles have been used to deliver the cells and

 

 14   been associated with different outcomes, and we

 

 15   don't know anything about biodistribution, and we

 

 16   really didn't look.

 

 17             So, what issues are there?  The issues are

 

 18   safety and efficacy obviously, and as we move up

 

 19   this continuum,  we can address these issues

 

 20   differently.  Safety obviously involves cells,

 

 21   delivery, dose, and we have to do that in relevant

 

 22   models.

 

 23             Efficacy involves the right model, acute

 

 24   MI potentially, looking at various cell types in a

 

 25   side-by-side way.  We have to begin to

 

                                                               264

 

  1   differentiate between diastolic heart failure.  I

 

  2   think the preclinical data that exists so far

 

  3   suggests that everything works to begin to improve

 

  4   remodeling and diastolic effects including cells

 

  5   that don't work in systolic heart failure.

 

  6             Systolic heart failure in our lab at

 

  7   least, we know fibroblasts don't work, stromal

 

  8   works less well than some other cell types, and I

 

  9   think the issues are open questions that have to be

 

 10   addressed.

 

 11             If you then believe that the issues effect

 

 12   are impact cells, delivery, and effect, how does

 

 13   that translate to the animal models?  Well, in

 

 14   small animals, I think we can ask questions about

 

 15   the cells.  We can ask questions about deriving the

 

 16   cells, we can ask questions about markers for the

 

 17   cells, we can ask in vitro questions.

 

 18             I think as we begin to move towards

 

 19   delivery and effect, we have to move up the animal

 

 20   continuum.  Delivery, we really can't do in small

 

 21   animal models, relevant delivery, we can't do.

 

 22   Effect, again, I don't think we can do in small

 

 23   animal models, we have to do it in larger animal

 

 24   models.

 

 25             In addition, when we start looking at the

 

                                                               265

 

  1   mechanism of effect, to some degree, if we are

 

  2   looking at angiogenesis, we can measure capillary

 

  3   density in small animals, but it is very difficult

 

  4   to get adequate measures of relevant vessels or

 

  5   perfused vessels in small animals, and I think we

 

  6   have to move up the continuum.

 

  7             In terms of myogenesis, we can get some

 

  8   data about ex vivo in isolated heart preparations

 

  9   and whether or not there are gross improvements in

 

 10   contractility.  We can begin to measure wall

 

 11   thickness, ejection fraction, and you can certainly

 

 12   do exercise studies in small animals and move up

 

 13   the continuum, but in terms of electrical

 

 14   compatibility and mechanical compatibility, you are

 

 15   never going to get it done in a rodent model, you

 

 16   have to do it in large animals.

 

 17             So, I think it is only relevant to work

 

 18   with small animals when you have no choice about

 

 19   the cells, but as you start to measure the

 

 20   important parameters of physiology, you have to

 

 21   move up the model.

 

 22             So, what are the possible effects of these

 

 23   cells?  I think they can cause unloading of the

 

 24   heart or reverse remodeling simply by altering the

 

 25   mechanical properties of the scar.  They could

 

                                                               266

 

  1   possibly engraft and become muscle and contribute

 

  2   to contraction.

 

  3             They can obviously potentially form

 

  4   vessels or secrete factors that recruit cells that

 

  5   improve blood flow. You know, if you think about a

 

  6   lot of the data we have seen so far, it is possible

 

  7   that the cells we are putting in are doing nothing

 

  8   but recruiting bone marrow derived cells and

 

  9   actually ramping up endogenous repair.

 

 10             It is possible that the cells have a

 

 11   paracrine effect and either change the cytokine

 

 12   supply to the scar or the remainder of the heart,

 

 13   or recruit other stem or progenitor cells.

 

 14             Another possibility that is not really

 

 15   talked about, and again you can do these studies in

 

 16   small animal models better than large animal

 

 17   models, is fusion with existing cardiocytes.  It is

 

 18   possible that the cells we inject actually fuse

 

 19   with cardiocytes that are hibernating and save

 

 20   those cells, and thereby contribute to contraction.

 

 21             If we are going to talk about the

 

 22   different possibilities, cell delivery and effect,

 

 23   and which animal models to use, I want to briefly

 

 24   say that we have to define our populations of

 

 25   cells, and that is easier in some animal models

 

                                                               267

 

  1   than others.

 

  2             Again, when you start looking at

 

  3   myoblasts, fibroblasts, and other cells, being able

 

  4   to do that in rodents is much easier than being

 

  5   able to do that in larger animals.  I think I have

 

  6   beat that horse, so I won't keep saying it, but

 

  7   cells, I think are studies we can do in rodents.

 

  8             You have seen this slide before where we

 

  9   begin to talk about how the cells might work and

 

 10   whether or not we have an effect on reverse

 

 11   remodeling or growing new cells, and whether or not

 

 12   we think we can do those studies in large animals

 

 13   or small animals.  I would submit that we are going

 

 14   to have to do those in large animals because we

 

 15   can't really measure remodeling and reverse

 

 16   remodeling in all of the new cells in the small

 

 17   animal models.

 

 18             Now, we took the approach that if we are

 

 19   going to start moving back up this cascade of

 

 20   events, that might, in fact, take a combination of

 

 21   angiogenesis and myogenesis, and it might take

 

 22   cells plus genes or multiple combinations of cells.

 

 23             Initially, we looked at cells plus genes

 

 24   by virally infecting myoblasts with VEGF

 

 25   adenovirus, and we measured the effect of those

 

                                                               268

 

  1   cells on capillary density in peripheral skeletal

 

  2   muscle and a hind limb ischemia model, and these

 

  3   are old data, so I am not going to spend much time

 

  4   on them other than to say that we found that

 

  5   myoblasts had a relatively significant effect in

 

  6   terms of increasing capillary density greater than

 

  7   VEGF virus alone or MT virus, but when we

 

  8   overexpress VEGF in the myoblasts, we got back up

 

  9   to about 75 percent of the control without the side

 

 10   effects that we saw with VEGF virus alone,

 

 11   angiomas.

 

 12             So, we decided to move into the

 

 13   myocardium, and these are data done with cells that

 

 14   overexpress another angiogenic factor where we

 

 15   looked at MRI, and I apologize, this is percent

 

 16   change and ejection fraction, and our historical

 

 17   controls, and these really are historical, they are

 

 18   not done at the same time, the active study was

 

 19   myoblasts versus angiogenic myoblasts versus

 

 20   historical shams.

 

 21             We found that we increased capillary

 

 22   density significantly in these animals.  These were

 

 23   studies done in mice.  We couldn't do these studies

 

 24   actually in rabbit because rabbit cells don't have

 

 25   the cell surface receptors to actually take up or

 

                                                               269

 

  1   be transfected with some of the viruses that we

 

  2   were trying to use, so that becomes a problem.

 

  3             But we believe that angiogenesis was more

 

  4   important than to promote cell survival or

 

  5   proliferation, and we also looked at Victor Dzaus'

 

  6   data, I wanted to show briefly.  You saw it when

 

  7   Silviu presented it, that if you overexpress a

 

  8   survival factor AKT in bone marrow stromal cells

 

  9   and transplanted them into the heart, survival

 

 10   increased.

 

 11             So, we know angiogenesis increases

 

 12   function, increasing survival increases function,

 

 13   so if angiogenesis helps and increasing survival

 

 14   helps, why use a gene, why not use a mixture of

 

 15   cells.

 

 16             We based that on data that we have gotten

 

 17   now in our hands where we were able to show that if

 

 18   we gave cells from young apoE animals, gave bone

 

 19   marrow cells from young apoE animals to apoE

 

 20   animals that were fed on a high fat diet, normally

 

 21   develop pretty bad atherosclerosis, that we could

 

 22   actually prevent this atherosclerosis.

 

 23             We took, we actually have now begun to

 

 24   take these cells and deliver them in combination

 

 25   with myoblasts to look at effects on function.  I

 

                                                               270

 

  1   don't have a slide, but I can tell you that we are

 

  2   beginning to see better effects on function with a

 

  3   combination of cells and with individual cells

 

  4   alone.  We couldn't do these studies except in

 

  5   mice, so there are times when mouse cells are

 

  6   relevant.

 

  7             We did another study where we began to

 

  8   look at bone marrow mononuclear cells.  These are

 

  9   data that were just presented at the ACC last week,

 

 10   where we actually infused mononuclear cells into

 

 11   the circulation of animals where we created a

 

 12   vascular injury.

 

 13             We are able to show in our sham-treated

 

 14   animals or animals treated with other cells that

 

 15   you had neointimal proliferation, that we were able

 

 16   to prevent with bone marrow mononuclear cells.

 

 17             So, again, we have now started the

 

 18   approach of delivering these cells in combination

 

 19   with myoblasts to see if we can have a more

 

 20   dramatic effect on not only myocardial repair, but

 

 21   on vascular repair, as well.

 

 22             So, what are the other factors that are

 

 23   likely involved that are going to affect the model

 

 24   we use?  The timing after injury, whether or not we

 

 25   can really grow old animals that replicate the six

 

                                                               271

 

  1   to seven years that are needed, the type of injury,

 

  2   it is not probably going to be feasible to do

 

  3   dilated cardiomyopathy studies in mice and inject

 

  4   significant numbers of animals, plus when we start

 

  5   trying to treat ischemic and chronic and acute

 

  6   animals, mice, the issue we have to consider is we

 

  7   have got a 1 mm infarct, we got a 1 mm infarct, and

 

  8   the cells are microns in diameter.

 

  9             Those cells are the same size essentially

 

 10   in rat, rabbit, pig, human, they are not much

 

 11   different in size, so the whole geometry of putting

 

 12   those cells in and getting an improvement is going

 

 13   to be much different than you are going to see in

 

 14   larger animal models.

 

 15             There are two issues that really don't

 

 16   affect what animal you choose, but they are not

 

 17   being discussed at all, and those are gender and

 

 18   age.

 

 19             Most of the preclinical data that we have

 

 20   published are in female rabbits, so we went back

 

 21   and started doing studies in male rabbits, and what

 

 22   we found is that male myoblasts die under

 

 23   conditions where female cells survive, and that is

 

 24   true both in vitro and in vivo, and that really

 

 25   surprised us, and we had to go back and begin to

 

                                                               272

 

  1   reevaluate what we think is going on here, and we

 

  2   are just beginning to follow up on that, but I

 

  3   think it is an interesting point that we are going

 

  4   to have to consider going forward.

 

  5             In terms of age, all of the studies we

 

  6   have done have been in old animals, but that is

 

  7   rarely the case.  It is rarely the case that old

 

  8   mice are used in these studies, that old pigs are

 

  9   used in these studies.  In fact, typically, people

 

 10   use young pigs because they want to keep them

 

 11   small, and don't want them to grow significantly

 

 12   over the duration of the studies.

 

 13             I think the numbers and kinds of cells

 

 14   that you can obtain are going to be very different.

 

 15   Other than cells, we have to consider the culture

 

 16   conditions, and we can't ignore the fact that

 

 17   autologous serum has been touted as one reason that

 

 18   there is a Spanish study where there aren't any

 

 19   abnormal electrical events even though there have

 

 20   been in all the other human studies.

 

 21             So, the injury models currently that we

 

 22   are using don't match the patients, and I think we

 

 23   are going to have to really think about that going

 

 24   forward.  We don't have heart failure models, we

 

 25   just don't.  Nobody is using heart failure models,

 

                                                               273

 

  1   but every patient is a heart failure patient.

 

  2             As I said before, I apologize, in terms of

 

  3   whether or not we can use small animals, I think

 

  4   for an isolated heart prep, mouse and rat are fine,

 

  5   but as you are going to start doing physiologically

 

  6   relevant studies, you have to move up the animal

 

  7   continuum, but there are limitations there, as

 

  8   well, so we have actually chosen, and I think more

 

  9   and more people are choosing, to use sonomicrometry

 

 10   or cine MRI.  Fortunately, you can use that for all

 

 11   of the animal models that have been proposed so

 

 12   far.

 

 13             I am not going to show those data.

 

 14             I actually want to end with two slides

 

 15   that show something that I have tried to gather

 

 16   from the clinical data that exists, but I think

 

 17   they make a point.

 

 18             We are talking about different animal

 

 19   models, but we are also talking about different

 

 20   cell types, and people are constantly saying how do

 

 21   myoblasts and bone marrow derived stem or

 

 22   progenitor cells compare.

 

 23             The point I want to make is they don't.

 

 24   If you look at the studies that exist so far--and

 

 25   these are clinical studies, not preclinical

 

                                                               274

 

  1   studies--if you look at myoblasts, the dose varies

 

  2   widely.

 

  3             If you look at bone marrow derived stem or

 

  4   progenitor cells, the dose is significantly less

 

  5   and the physiologically relevant cells are a very

 

  6   small subset of those, as you just heard.

 

  7             Moreover, the patients differ greatly.

 

  8   With myoblasts, the patients are from greater than

 

  9   one month to end-stage heart failure, but with the

 

 10   progenitor cells, the patients are 3 to 9 days

 

 11   post-MI or have refractory angina.

 

 12             The delivery methods differ significantly.

 

 13   They are intracardiac for myoblasts, surgically or

 

 14   percutaneously.  They are intracoronary for the

 

 15   bone marrow derived cells.  Yet, people are trying

 

 16   to compare the outcomes from these, and I think

 

 17   that is true, not just clinically, but

 

 18   preclinically, as well, as people are trying to

 

 19   make the argument for their cell type.

 

 20             So, until we are doing side-by-side

 

 21   studies with the same cell type and the same animal

 

 22   model, I really think we can't draw conclusions

 

 23   about what is going on.

 

 24             So, the questions that I think are really

 

 25   out there are:  Is there a best cell?  I don't

 

                                                               275

 

  1   think we know that there is yet.

 

  2             Should we use just growth factors?  I

 

  3   think the GCSF data would suggest not yet, we don't

 

  4   understand it well enough.

 

  5             Is there a future for biologic devices?

 

  6   Probably. Are we going to be putting cells on

 

  7   stents?  I don't know, but we probably are going to

 

  8   be putting them on patches and other devices in the

 

  9   very near future.

 

 10             There is a real question, dose, timing,

 

 11   and patients, probably, but our models need to

 

 12   mimic that.

 

 13             I will stop there.

 

 14             [Applause.]

 

 15                               Q&A

 

 16             DR. RAO:  Thank you, Doris.  I wanted to

 

 17   start off by asking you one question first.  That

 

 18   was that implicit in all of these talks throughout

 

 19   has been the fact that it seems to be important

 

 20   when choosing a model that you have to have the

 

 21   right markers.  That really is because you are

 

 22   doing syngeneic transplants.  You want to put the

 

 23   same animal species cells back into the animal to

 

 24   do the follow-up.

 

 25             Nobody, at least in this field, seems to

 

                                                               276

 

  1   think that you can immunesuppressed mouse and use

 

  2   human cells directly, or do some equivalent

 

  3   otherwise, and that that is reasonable.

 

  4             Now, is that true?  That is important

 

  5   because this is going to be really important in the

 

  6   future. I just wanted to get your feel for that.

 

  7             DR. TAYLOR:  I think that is fairly

 

  8   accurate, and I think the problem has been that we

 

  9   haven't had the opportunity to really measure the

 

 10   effects of these cells in mice very well until the

 

 11   last couple of years.

 

 12             Until the last probably year and a half to

 

 13   two years, you couldn't do MRI in a mouse reliably.

 

 14   You certainly couldn't get a good enough image to

 

 15   measure regional versus global function.

 

 16             Sonomicrometry was hard to do in a mouse.

 

 17   The pressure volume catheters weren't quite up to

 

 18   snuff, and so the ability to make those

 

 19   measurements weren't true. Moreover, most of us

 

 20   believed early on at least that autologous cells

 

 21   were more likely to be clinically accepted, and

 

 22   patients certainly liked the concept of getting

 

 23   their own cells better than the concept of getting

 

 24   somebody else's.

 

 25             So, I think we chose those cells because

 

                                                               277

 

  1   they made sense to us clinically, and we were

 

  2   technically limited by our ability to make the

 

  3   measurements with other cells.

 

  4             In terms of using human cells in

 

  5   immunocompromised rodents, I think we can do that

 

  6   now, but whether or not--I think it is an open

 

  7   question about whether or not we are going to get

 

  8   the best functional outcomes.

 

  9             DR. ITESCU:  Can I just maybe add a little

 

 10   bit to that?  You know, we use the

 

 11   immunocompromised rodents pretty well, but I think

 

 12   what we are learning as we move forward is that

 

 13   even the so-called immunocompromised rodents are

 

 14   not fully immunocompromised, and you have really

 

 15   got to start understanding which kind of lineages

 

 16   in their immune system remain active, and what

 

 17   impact does that have on the cells you are putting

 

 18   in.

 

 19             We are now at the point where we are

 

 20   adding cocktails to try to remove even the residual

 

 21   immune function in these kind of animals.

 

 22             On the other hand, I think that if you are

 

 23   trying to use a cocktail of immunosuppressive

 

 24   agents in a normal animal, I think then you are

 

 25   going to run into the problems of what effects all

 

                                                               278

 

  1   these drugs have on the cells that we are putting

 

  2   in, are they inhibiting differentiation, are they

 

  3   inhibiting function.  There is a whole range of

 

  4   issues that I don't think we want to get into.

 

  5             DR. RAO:  Absolutely, that is important.

 

  6             DR. HARLAN:  I will make a comment on

 

  7   that, and then I had a question.  I agree, in the

 

  8   islet transplant field that I know best, for

 

  9   instance, in order to correct a mouse with human

 

 10   islets, you need about 1,500 islets, whereas, you

 

 11   need about 400 rodent islets, and presumably it is

 

 12   because there are species differences in the

 

 13   factors that support the growth of those cells.

 

 14             Then, I agree and appreciate your talk,

 

 15   but I would extend it in two ways.  I think a theme

 

 16   of your talk was that the large animal models in

 

 17   general tend to be better than rodents in

 

 18   predicting things, and I think that is true, but I

 

 19   wish to point out that all models are models.

 

 20             In our transplant studies, we did things

 

 21   in primates, testing various immunotherapies in

 

 22   different systems, and it worked beautifully and

 

 23   failed miserably in the clinic, so even large

 

 24   animal models, even using nonhuman primates, are

 

 25   models, and they have variables that are hard to

 

                                                               279

 

  1   predict.

 

  2             DR. TAYLOR:  I would add one comment to

 

  3   that, which is all of us are looking at progenitor

 

  4   cells and the effects of these progenitor cells on

 

  5   cardiovascular function, and we know that the

 

  6   animals we are treating are relatively acutely ill

 

  7   even if we have given them heart failure, but

 

  8   patients are very ill.

 

  9             They have had things impacting their

 

 10   progenitor cells for years, that we know nothing

 

 11   about, and it is not just aging, it's drugs, it's

 

 12   other things, so the cells we get out and the cells

 

 13   we put back in are going to be very different than

 

 14   cells we get out and put back in, in animals that

 

 15   we have only made sick for a month or a year.

 

 16             DR. HARLAN:  And because we don't know all

 

 17   the factors that made people sick, generating that

 

 18   perfect animal model may be an impossible ideal,

 

 19   that's the only point, I think we are agreeing.

 

 20             The second thing is, though, that you

 

 21   didn't mention is in any model, and when you go to

 

 22   the clinic, I think it is important and we started

 

 23   the day that way, identifying the patients for whom

 

 24   existing therapies just have failed, so that is one

 

 25   way where you can go.  When you have got nothing

 

                                                               280

 

  1   else to offer, and you have the patients in a bad

 

  2   position, then, I think it passes the threshold.

 

  3             DR. TAYLOR:  But those aren't the models

 

  4   that people are using.

 

  5             DR. HARLAN:  Well, we will talk about that

 

  6   tomorrow, I agree.

 

  7             Then, the third point, I say this in jest,

 

  8   and I don't buy it, but I like your Ghandi quote,

 

  9   and I will cite another philosopher, W.C. Fields,

 

 10   who said, "If at first you don't succeed, try

 

 11   again; if it fails again, you might as well give

 

 12   up, there is no sense being a damn fool about it."

 

 13             I don't agree with that, it's just the

 

 14   other take.

 

 15             DR. RAO:  Dr. Epstein.

 

 16             DR. EPSTEIN:  I just wanted to make a

 

 17   point, which I think is important because I sense

 

 18   that one could very easily come to the conclusion

 

 19   without thinking it through further that a large

 

 20   animal model is the really only valid preclinical

 

 21   model.

 

 22             I think Doris sort of made this point, but

 

 23   it might have been lost.  It depends on what you

 

 24   are looking at.  For example, if are interested in

 

 25   myogenesis, I would agree, ultimately, you have to

 

                                                               281

 

  1   go to the large animal model because the small

 

  2   rodents, mice, become difficult to draw conclusions

 

  3   about, but if you are looking at angiogenesis, the

 

  4   mouse model is much better, I think, than a large

 

  5   animal model.

 

  6             We have excellent ways of measuring

 

  7   perfusion now, superb ways of measuring perfusion,

 

  8   and what comes up in our laboratory a lot is if we

 

  9   give an intervention that has an effect, and then

 

 10   we want to see whether we could further enhance

 

 11   that, like stromal cells and then genetic

 

 12   engineering of stromal cells, in a pig model of

 

 13   myocardial ischemia, it starts out with 85 percent

 

 14   of normal.

 

 15             If you get it up to 95 percent of normal

 

 16   with your first intervention, you have no room to

 

 17   look at the next step, whereas, with mice, that

 

 18   could be modulated much more easily, and we are

 

 19   able to demonstrate a primary effect and then an

 

 20   additional effect on it.

 

 21             So, I think, you know, I wouldn't like the

 

 22   FDA to go away with the conclusion that you have to

 

 23   do an efficacy model in a pig or a dog to go to the

 

 24   clinic.  Again, it depends what you are looking at.

 

 25             I will make a point about the

 

                                                               282

 

  1   immunosuppressed animals.  Angiogenesis, an

 

  2   intrinsic component of the angiogenic process is

 

  3   inflammation, so if you put in cells in a model

 

  4   that is immunocompromised, it is a laboratory model

 

  5   where you have taken away one of the normal

 

  6   modulating influences, so you have to be very

 

  7   careful using an immunocompromised model at least

 

  8   to look at a process in which inflammation is a

 

  9   very critical component.

 

 10             DR. RAO:  A point well taken.

 

 11             Bruce.

 

 12             DR. BLAZAR:  Doris, I wanted to ask you,

 

 13   since you have reviewed all of the preclinical

 

 14   data, and you had the tenet that the data should

 

 15   drive the studies, what of the preclinical data is

 

 16   sufficiently compelling that this would have driven

 

 17   the studies to go forward, are the models done so

 

 18   far incomplete to be able to decide on appropriate

 

 19   studies?

 

 20             I know we are going to talk about that

 

 21   tomorrow, but since you have reviewed in two talks

 

 22   this issue--

 

 23             DR. TAYLOR:  I think there is certainly a

 

 24   lot more preclinical data from myoblasts than there

 

 25   are for some of these bone marrow derived

 

                                                               283

 

  1   progenitor cells.  I think that is in part because

 

  2   we have known about myoblasts since 1961 and we

 

  3   have know about these bone marrow derived cells for

 

  4   the last five or six years, so it is not

 

  5   surprising.

 

  6             I think myoblasts have been used in

 

  7   peripheral models, they have been used in cardiac

 

  8   models of injury, they have been used in large

 

  9   animals, they have been used in small animals, and

 

 10   there is a confluence of data, all of which say if

 

 11   you give these cells, the animals get better.

 

 12             I think most of the cardiac models were

 

 13   relatively acutely after injury, within a month

 

 14   after injury, but nonetheless, they said if you use

 

 15   these cells, the animals get better.

 

 16             I think what is starting to happen in the

 

 17   bone marrow mononuclear cell field and in the bone

 

 18   marrow stromal cell field, and in even the MAPC

 

 19   field, is that we are seeing isolated studies with

 

 20   cells that are called a given thing, but aren't

 

 21   necessarily defined the same way.

 

 22             The way one group defines an EPC, and the

 

 23   way another group defines an EPC may be very

 

 24   different, so there is not necessarily a confluence

 

 25   of data yet, and you can't necessarily even compare

 

                                                               284

 

  1   some of the preclinical studies because the cells

 

  2   are very different, or we don't know what the

 

  3   criteria are for those cells.

 

  4             So, I think that what has to happen is we

 

  5   have to know what the definition of the cells are,

 

  6   and that is only now becoming true for endothelial

 

  7   progenitor cells.

 

  8             DR. BLAZAR:  So, a corollary of this

 

  9   question is, is it pretty well established by those

 

 10   of you in the field, what the bar is that you have

 

 11   to get over.

 

 12             It seems the way you have described this,

 

 13   it's a systolic function bar that is--no.

 

 14             DR. TAYLOR:  I think it depends on what

 

 15   you believe the mechanism of action is for the

 

 16   cells, and I think again we don't know that, but it

 

 17   looks like what is coming about generally is that

 

 18   for bone marrow derived cells, the goal may be

 

 19   angiogenesis, and for muscle-derived cells, the

 

 20   goal may be myogenesis, and those have different

 

 21   criteria and different preclinical studies that I

 

 22   think you ought to do.

 

 23             DR. BLAZAR:  But that presumes for

 

 24   clinical applications that you know exactly the

 

 25   pathophysiology of the lesion you are trying to

 

                                                               285

 

  1   treat.

 

  2             DR. TAYLOR:  I think it begins to argue

 

  3   for timing after injury.

 

  4             DR. BLAZAR:  Okay.

 

  5             DR. RAO:  If anybody has a really brief

 

  6   comment, otherwise, we will take Dr. Mul and then

 

  7   Dr. Schneider.

 

  8             Do you have a quick comment?

 

  9             DR. MENASCHE:  Go ahead.  I just had a

 

 10   comment, but I can wait.

 

 11             DR. MULE:  I would just like to follow up

 

 12   on Bruce's questions.  This has been an incredibly

 

 13   frustrating afternoon for me from the standpoint

 

 14   that I think the presentations have nicely pointed

 

 15   out the strengths and weaknesses of small animal

 

 16   models versus large animal models.

 

 17             Overlaid on that is the fact that none of

 

 18   these models really are good models for the actual

 

 19   disease state in humans.  What I was hoping to hear

 

 20   this afternoon was taking the strengths and

 

 21   weaknesses of each of the models and maybe laying

 

 22   out, hopefully tomorrow, what an ideal, if we could

 

 23   go that way, what the ideal recommendations would

 

 24   be for the field to help the clinicians, such as

 

 25   Philippe, in conducting the next generation trials.

 

                                                               286

 

  1             That is the real concern to me, is that at

 

  2   the end of the day, the horse is out of the gate,

 

  3   and Philippe and others are going to be conducting

 

  4   these trials rather rapidly, and I think we need to

 

  5   help them to establish some guidelines as to

 

  6   whether or not we should abandon animal models,

 

  7   move on to the clinic, and design the clinical

 

  8   trials in such a way that we get the best

 

  9   scientific data available and the best clinical

 

 10   situations that can be defined, and define the

 

 11   endpoints of the clinical trial with the

 

 12   appropriate placebo.

 

 13             So, I just wanted to lay out that my

 

 14   deepest concern is that years from now, we will be

 

 15   using the same models that you have very nicely

 

 16   summarized, small animals, large animals, fully

 

 17   aware that these limitations continue to exist, and

 

 18   whether or not those data that are generated over

 

 19   the next several years will help Philippe and

 

 20   others to characterize how we should go forward in

 

 21   conducting these clinical trials.  It is just some

 

 22   comments I had.

 

 23             DR. RAO:  I think you are echoing what the

 

 24   FDA is feeling, I guess, right now.

 

 25             Dr. Schneider.

 

                                                               287

 

  1             DR. SCHNEIDER:  Doris, you know that I

 

  2   share your general cautionary note and share many

 

  3   of the specifics, but let me disagree with two

 

  4   specific points that you made about the lack of

 

  5   utility or impediments to the use of the mouse.

 

  6             One of them that you singled out is the

 

  7   impediment to using the mouse as a model for

 

  8   studying the electrical connectivity of donor cells

 

  9   to the host environment, which has become an issue

 

 10   of prominence because of the issue of ventricular

 

 11   tachycardia.

 

 12             I think Loren Field has shown very

 

 13   convincingly you can use 2-photon microscopy to

 

 14   study the propagation of action potentials on the

 

 15   epicardial surface, you can study the propagation

 

 16   of calcium transients in the mouse heart with

 

 17   grafted cells, so that is a non-issue as of this

 

 18   year.

 

 19             DR. TAYLOR:  I completely disagree, but we

 

 20   can have that discussion--

 

 21             DR. SCHNEIDER:   Perhaps so, but

 

 22   Circulation doesn't, and there is adequate

 

 23   peer-reviewed data out there that shows that it is

 

 24   technically feasible with some esoteric

 

 25   instrumentation.

 

                                                               288

 

  1             The same thing I think is true in terms of

 

  2   the issue of mechanics.  As you say yourself, cine

 

  3   MRI levels the playing field across all of these

 

  4   species, and at a few centers, cine MRI is even

 

  5   being combined with spam, so that people can do

 

  6   finite element analysis in the small mammals.

 

  7             So, to me, and I share Dr. Mul's

 

  8   frustration, I tend to focus myself on a different

 

  9   issue, and that is not whether the small mammal is

 

 10   adequately achievable for the endpoints that we

 

 11   want to study, but is the small mammal adequately

 

 12   predictive of the pathophysiology we want to study.

 

 13             There, the issues include the fact that

 

 14   neither the small nor the large mammal is done in

 

 15   an aged animal against a background of diffuse

 

 16   atherosclerotic disease.

 

 17             DR. TAYLOR:  I want to just comment on

 

 18   that looking at electrical connectivity is not the

 

 19   same thing as looking at ventricular tachycardia or

 

 20   arrhythmias, and that it is going to be difficult

 

 21   to do those studies when the heart is beating at

 

 22   600 beats per minute, and that is I think one of

 

 23   the main issues, as well as the fact that most

 

 24   electrophysiologists tell you that the larger

 

 25   geometry is much more conducive, of the human

 

                                                               289

 

  1   heart, is much more conducive to electrical

 

  2   abnormalities than the smaller geometry of a rodent

 

  3   heart, so you are not going to see the same

 

  4   properties even if you have an injury there.

 

  5             DR. SCHNEIDER:  Again, it becomes an issue

 

  6   of whether it is a predictive biology, not an issue

 

  7   of whether it is technically achievable.

 

  8             DR. TAYLOR:  Sure.

 

  9             DR. MENASCHE:  To some extent, Dr. Mul

 

 10   has anticipated my comment.  What I wanted to

 

 11   emphasize from a clinical standpoint is that

 

 12   regardless of the animal model we are going to

 

 13   use--and I have advocated large animal models, as

 

 14   well--there is not a single animal preparation

 

 15   which can realistically model the very complex

 

 16   situation of the patients we are dealing with.

 

 17             There is not a single model which can

 

 18   reproduce the situation of a 70-year-old person

 

 19   with Class III heart failure, two previous bypass,

 

 20   seven angiopathies, collaterals, and so on, so at

 

 21   what point we really have to be prepared to move

 

 22   forward and to go across the gap.

 

 23             Now, considering skeletal myoblasts, we

 

 24   have clear evidence regardless of the limitations

 

 25   of the models that the technique can be implemented

 

                                                               290

 

  1   easily in patients.

 

  2             Number two, we have reasonable evidence

 

  3   that it is safe provided some precautions are

 

  4   taken.  We don't fully understand the mechanisms,

 

  5   but many interventions are currently using patients

 

  6   without an extensive understanding of the

 

  7   mechanisms.  I think it took years before people

 

  8   understood how aspirin was working.

 

  9             So, as we are continuing to try to

 

 10   understand the mechanisms, I think that given the

 

 11   huge population we have to deal with, and the

 

 12   number of patients without any option, it is

 

 13   critically important and timely appropriate now to

 

 14   move to the efficacy studies, and we are certainly

 

 15   looking for advice and help for designing the study

 

 16   in such a way that they can draw meaningful

 

 17   conclusions and answers to the two fundamental

 

 18   questions, does it improve function in the areas

 

 19   where cells are put, does this improvement have an

 

 20   impact on the clinical outcomes.

 

 21             DR. TAYLOR:  Philippe, the one comment I

 

 22   would make to that is I think you are absolutely

 

 23   right, we don't have good animal models, I don't

 

 24   anyone in the room will argue that.

 

 25             I think the thing I would add to what you

 

                                                               291

 

  1   said is you mentioned no-option patients, but those

 

  2   aren't the patients that are involved in some of

 

  3   the studies that are going forward, and I think

 

  4   those are the patients who should be involved in

 

  5   the studies going forward, and I hope that is one

 

  6   of the things that will emerge from this, that it

 

  7   may be responsible to go forward in those groups of

 

  8   patients, it may not be responsible to go forward

 

  9   in some other groups without more preclinical data.

 

 10             DR. RAO:  I would like to just remind

 

 11   everyone that part of the discussion is not

 

 12   specific to one particular cell type, but is in

 

 13   general, and maybe the conclusion may be that it is

 

 14   just simply we have to have differing criteria

 

 15   depending on the cell type or the model, or

 

 16   whatever, when we discuss it, at least its options,

 

 17   and that may be as far as one can go.

 

 18             So, keep that thought in mind, that

 

 19   nothing that  you hear necessarily means this is

 

 20   absolute or anything in that fashion.

 

 21             DR. ITESCU:  Along those lines, I think

 

 22   the point  you are making is exactly right.  Some

 

 23   cell products are based on characterization of the

 

 24   surface phenotype, based on many years of

 

 25   immunoselection with markers only present in humans

 

                                                               292

 

  1   and not present in any other species.

 

  2             Some cell types have been characterized by

 

  3   very different biologists, who have used functional

 

  4   outcomes and functional criteria.  So, you are

 

  5   talking about apples and oranges really, so you

 

  6   can't have the same clinical models or preclinical

 

  7   models for those, so you have got to define

 

  8   appropriate models for each of those cell types,

 

  9   because they have been characterized from totally

 

 10   different perspectives.

 

 11             DR. RAO:  I thought we might use that to

 

 12   start the conversation tomorrow, yes.

 

 13             Everybody looks like they need a break, so

 

 14   we will take a short 10-minute break.

 

 15             [Break.]

 

 16             DR. RAO:  Welcome back, everybody.

 

 17             We are going to change topics a little bit

 

 18   and talk about devices now.  It is finally going to

 

 19   be the turn of Dr. Jensen to talk on cardiac

 

 20   catheters.

 

 21        Cardiac Catheters for Delivery of Cell Suspensions

 

 22             DR. JENSEN:  I am going to go ahead and

 

 23   get started because we are running a little bit

 

 24   slow here, and I am also going to try and see if I

 

 25   can catch up a little bit in terms of time.

 

                                                               293

 

  1             My name is Nick Jensen.  I work in the

 

  2   Division of Cardiovascular Devices in FDA's Center

 

  3   for Devices and Radiological Health, and I have

 

  4   been asked to briefly introduce the cardiac

 

  5   catheters that have been used for delivery of cell

 

  6   therapies intended to treat cardiac disease.

 

  7             My presentation today will be limited to

 

  8   potential questions that relate to the interaction

 

  9   between cell therapy suspensions and the cardiac

 

 10   catheters used to deliver these therapies.

 

 11             Further, the questions that I list today

 

 12   are among the standard questions that we currently

 

 13   suggest to all sponsors either of cell therapy INDs

 

 14   or for these investigational catheters.

 

 15             As examples, we will discuss two types of

 

 16   cardiac catheters that have been used to deliver

 

 17   cell therapies to the heart.

 

 18             First, is infusion of cells into a

 

 19   coronary artery during a balloon occlusion of the

 

 20   artery.  Second, is needle-tipped injection

 

 21   catheters designed to permit percutaneous

 

 22   transendocardial injection into the myocardium.

 

 23             Again, these simply represent the methods

 

 24   and catheter types that have been most commonly

 

 25   used to date,  and they are again meant to provide

 

                                                               294

 

  1   useful examples for discussion.  I think it is hard

 

  2   to predict what types of devices, what delivery

 

  3   methods may be used in the future.

 

  4             The first method, infusion of cells into a

 

  5   coronary artery offers potential advantages that

 

  6   include simplicity and ease of use.  As has been

 

  7   mentioned earlier today, this method may not be

 

  8   suitable for all types of cell suspensions.

 

  9   Potential limitations include the potential

 

 10   requirement that infused cells be able to migrate

 

 11   from the vasculature into the myocardium, and a

 

 12   potentially increased risk of embolization or

 

 13   microembolization if some types of cells are

 

 14   infused using this method.

 

 15             Published case series using this method

 

 16   have demonstrated preliminary clinical feasibility

 

 17   both when used within hours to days following acute

 

 18   myocardial infarction, and this is often following

 

 19   emergency stent placement at the site of thrombotic

 

 20   occlusion.

 

 21             It has also been used in patients who

 

 22   suffer from chronic myocardial infarction and

 

 23   ischemia.

 

 24             In the studies reported to date, balloon

 

 25   catheters have typically been used to temporarily

 

                                                               295

 

  1   occlude the coronary artery proximal to the

 

  2   treatment region.  The desired cell suspension is

 

  3   then infused into the artery distal to the inflated

 

  4   balloon either using a lumen within the balloon

 

  5   catheter, typically, a guidewire lumen, or using a

 

  6   separate infusion catheter placed lateral to the

 

  7   balloon catheter, such that it lies between the

 

  8   inflated balloon and the artery wall.

 

  9             Use of balloon occlusion permits infusion

 

 10   of cells at pressures that exceed coronary artery

 

 11   pressure, and it has been hypothesized that

 

 12   increased infusion pressures may provide benefits

 

 13   that include increased dispersion of cells within

 

 14   the vasculature, increased adhesion of cells to the

 

 15   vascular endothelium, and increased migration of

 

 16   cells across the vascular endothelium and into the

 

 17   myocardium.

 

 18             This familiar illustration from a

 

 19   publication by Strauer, et al., illustrates the use

 

 20   of a coronary artery balloon catheter to infuse

 

 21   cells into a region of acute myocardial infarction.

 

 22             In this illustration, the balloon catheter

 

 23   has been inserted into a large artery, directed

 

 24   retrograde through the aorta, then, into a coronary

 

 25   artery, and then directed distal within that artery

 

                                                               296

 

  1   to the site of acute thrombosis.

 

  2             Following inflation of the balloon to

 

  3   obstruct the artery, a syringe is used to infuse

 

  4   cell suspension through the guidewire lumen of this

 

  5   catheter and into the coronary artery distal to the

 

  6   balloon.

 

  7             Finally, although it is not obvious in

 

  8   this illustration, in this clinical study, the

 

  9   balloon was inflated at the site of acute

 

 10   thrombosis, and more specifically, at a site where

 

 11   a coronary artery stent was placed as an emergency

 

 12   treatment for the myocardial infarction, and that

 

 13   is potentially important because balloon deployment

 

 14   within a coronary artery stent can largely protect

 

 15   the artery from one potential concern that we will

 

 16   discuss briefly today, and that is the potential

 

 17   for damage to the artery caused by balloon

 

 18   inflation and subsequent stretching of the artery.

 

 19              Studies of this cell delivery method

 

 20   reported to date have commonly used balloon

 

 21   angioplasty catheters to occlude the artery.  The

 

 22   catheters are originally designed to stretch the

 

 23   lumen of an occluded fibrotic atherosclerotic

 

 24   coronary artery to a specific diameter that has

 

 25   been selected in advance by the treating physician.

 

                                                               297

 

  1             They are also used to expand coronary

 

  2   artery stents and again to prespecify diameter.

 

  3   Although these catheters were not originally

 

  4   designed for occlusion of an artery, they can be

 

  5   used for that purpose, followed by cell suspension

 

  6   and, additionally, as noted, if these catheters

 

  7   have a central guidewire lumen, that lumen can then

 

  8   potentially be used to infuse the cell suspension.

 

  9             Potential considerations when you are

 

 10   using an angioplasty catheter for this purpose

 

 11   include the following. First, contact between the

 

 12   catheter lumen materials and the cell suspension

 

 13   can potentially adversely affect either the

 

 14   viability or the functionality of the infused

 

 15   cells.

 

 16             Additionally, those cells may contact

 

 17   various lubricants that are commonly applied to the

 

 18   guidewire lumens during manufacturing for the

 

 19   purpose of facilitating guidewire passage.

 

 20             As a note, we are not aware yet of

 

 21   published reports that have examined whether

 

 22   catheter lumen materials may adversely affect

 

 23   viability or functionality of cells, however, this

 

 24   published animal study cited in the slide evaluated

 

 25   cardiac delivery of a transgene by an adenovirus

 

                                                               298

 

  1   vector.

 

  2             The investigators found that some lumen

 

  3   materials tested for use in a prototype needle

 

  4   injection catheter, the second type of catheter we

 

  5   will discuss today, adversely affected both the

 

  6   viral activity and viral transduction.

 

  7             They also found that a change in lumen

 

  8   materials was sufficient to completely eliminate

 

  9   these adverse side effects.

 

 10             Additional considerations.  Number two.  A

 

 11   second consideration when balloon angioplasty

 

 12   catheters are used for this purpose, infusion of

 

 13   cells, is that the balloon was originally designed

 

 14   to stretch a coronary artery, in other words, to

 

 15   controllably damage the artery, and for this use,

 

 16   it must instead be used to occlude that artery,

 

 17   hopefully, without damaging it.  This is

 

 18   potentially important.

 

 19             The degree of artery wall stretch that is

 

 20   typically created during balloon angioplasty will

 

 21   also subsequently induce arterial stenosis due to

 

 22   multiple mechanisms.  We think it is therefore

 

 23   essential that safe methods for balloon inflation

 

 24   be developed and demonstrated if you want to use an

 

 25   angioplasty catheter for this purpose.

 

                                                               299

 

  1             Importantly, development of safe methods

 

  2   for this new use could be complicated because

 

  3   balloon angioplasty catheters have widely varying

 

  4   pressure diameter relationships, in other words,

 

  5   compliance can vary greatly between different

 

  6   catheter models, so therefore the methods that are

 

  7   developed for one catheter may not be applicable to

 

  8   another one.

 

  9             A third potential concern related to this

 

 10   is the potential that concentrated cell suspensions

 

 11   may clog the catheter lumen.

 

 12             A fourth concern is that the lumens and

 

 13   connectors of angioplasty catheters are primarily

 

 14   designed for passage of a guidewire.  They may not

 

 15   have been tested for the ability to sustain high

 

 16   pressures that can occur during infusion of

 

 17   concentrated cell suspensions.

 

 18             The second type of catheter we will

 

 19   discuss today are needle-tipped injection

 

 20   catheters.  This method of cell delivery also

 

 21   offers potential advantages.  Notably, first, the

 

 22   ability to directly inject cells into desired

 

 23   myocardial locations.  Second, the potential for

 

 24   use with all types of cells.

 

 25             The investigational cell delivery systems

 

                                                               300

 

  1   developed for this therapy consists either of a

 

  2   catheter or of a system comprised of a catheter

 

  3   plus delivery sheaths, that include a retractable

 

  4   distal injection needle.  None are approved

 

  5   currently for sale in the U.S., however when we

 

  6   consider these new devices, it may be useful to

 

  7   note that some design requirements, and thus,

 

  8   potentially catheter characteristics, may be

 

  9   similar to requirements for other currently

 

 10   marketed cardiac catheters, potentially including

 

 11   both cardiac electrophysiology ablation catheters

 

 12   and endocardial biopsy catheters.

 

 13             More specifically, all three types of

 

 14   catheters or catheters plus sheaths would generally

 

 15   require a steerable or deflectable tip in order to

 

 16   facilitate direction of the catheter tip to various

 

 17   endocardial locations, and all three types of

 

 18   catheters must be sufficiently stiff to permit the

 

 19   user to maintain stable contact between the

 

 20   catheter tip and the moving endocardial surface of

 

 21   the ventricle.

 

 22             These illustrations are from a publication

 

 23   by Dr. Perin's group, and they illustrate one,

 

 24   investigational needle-tipped injecting catheter.

 

 25   The photo on the left illustrates a complete

 

                                                               301

 

  1   catheter including controls on the catheter handles

 

  2   that deflect the tip and that extend and retract

 

  3   the needle.

 

  4             The needle would normally be retracted

 

  5   into the catheter except when briefly extended for

 

  6   each injection that is made during a therapy

 

  7   session, and the syringe, potentially loaded with

 

  8   therapy suspension, is attached to an infusion

 

  9   port, also on the catheter handle.

 

 10             The drawing on the right illustrates

 

 11   delivery of the catheter retrograde through the

 

 12   aortic arch, then, through the mitral valve, and

 

 13   into the left ventricular cavity.  In this drawing,

 

 14   as you see, the catheter tip is deflected and the

 

 15   catheter is being used to make multiple injections

 

 16   from the endocardial surface of the ventricle.

 

 17             There are also potential concerns that

 

 18   attach to the use of needle injection catheters for

 

 19   delivery of cell therapies for cardiac disease.

 

 20             First, we believe this type of catheter

 

 21   may potentially be particularly prone to clogging

 

 22   by cell suspensions.  Factors that might contribute

 

 23   to this include the following:

 

 24             The potential desirability of using very

 

 25   small injection volumes plus highly concentrated

 

                                                               302

 

  1   cell suspensions, the potential desirability of a

 

  2   small diameter injection needle that will also,

 

  3   thus, have a small diameter lumen, and the fact

 

  4   that injections may be made at more than 20

 

  5   locations during a treatment session, thus,

 

  6   increasing the potential for clogging due to the

 

  7   repeat injections.

 

  8             A second concern.  As noted previously, it

 

  9   may be important to ask whether cell viability or

 

 10   functionality could be adversely affected by

 

 11   contact with catheter lumen materials.  In this

 

 12   type of catheter particularly, we think it may also

 

 13   be important to ask whether shear forces produced

 

 14   during infusion of cells through a long,

 

 15   potentially a very small diameter injection lumen

 

 16   might also adversely affect the cells.

 

 17             Third, it may be important to consider the

 

 18   safety, and this was brought up in one earlier

 

 19   session, of whether the safety of the cell

 

 20   suspension is accidentally delivered in the

 

 21   systemic circulation.

 

 22             Of note, with this type of catheter, it

 

 23   may be very difficult to maintain continuous

 

 24   contact between the catheter tip and the moving

 

 25   endocardial surface of the beating heart, and when

 

                                                               303

 

  1   contact is broken, therefore, you could have

 

  2   injection into the ventricular cavity and into the

 

  3   circulation.

 

  4             There is a note, for folks in the

 

  5   audience, I don't know if you can see the

 

  6   reference, but if I understand correctly, we will

 

  7   have cells on the web site, is that correct?  Okay.

 

  8   Otherwise, I was going to read the reference to

 

  9   you.

 

 10             One note, if people want to look into

 

 11   this, this has been studied for cardiac ablation

 

 12   catheterization electrophysiology.  I have noted

 

 13   one good study up here where they used intracardiac

 

 14   ultrasound imaging catheters to evaluate the

 

 15   difficulty of maintaining continuous contact when

 

 16   they thought contact was perfect.

 

 17             A fourth consideration.  Should we assume

 

 18   that it is important  to control or limit the

 

 19   maximum needle extension?  Factors to consider

 

 20   might include the following: Is it critical to

 

 21   avoid injection or laceration of the organs that

 

 22   surround the heart?

 

 23             It may also be important to consider

 

 24   whether there may be safety concerns if the cell

 

 25   suspension is inadvertently injected into the

 

                                                               304

 

  1   pericardial or thoracic spaces, or if it is drained

 

  2   from these spaces by the lymphatics and then

 

  3   delivered into the systemic circulation. Relating

 

  4   to needle injection, curves or bends in many

 

  5   catheter designs, including the 180-degree bend

 

  6   around the aortic arch that will normally be

 

  7   present, could affect the needle extension length

 

  8   of the catheter.

 

  9             Finally, particularly in hearts that have

 

 10   minimal epicardial fat surrounding the left

 

 11   ventricle, it may be difficult to avoid occasional

 

 12   injection completely through the wall of the

 

 13   ventricle and into the pericardial space.

 

 14             Factors that might contribute to this

 

 15   could include the following:

 

 16             First, is locally thin regions in the

 

 17   ventricle, possibly related to myocardial

 

 18   infarction, possibly related to the normal

 

 19   indentations that separate the muscular trabeculae

 

 20   of the ventricle on the endocardial surface of the

 

 21   heart.

 

 22             Second, is compression or stretching of

 

 23   the ventricular wall where the catheter tip is

 

 24   again pressed into contact with the wall, and,

 

 25   finally, the possibility that a forceful injection

 

                                                               305

 

  1   could simply potentially separate both myocardial

 

  2   cells and epicardial cells, allowing cell

 

  3   suspension to flush completely through the

 

  4   ventricular wall.

 

  5              A fifth and final question regarding

 

  6   needle injection catheters is the following:  Are

 

  7   injection depth and the spread of injection of the

 

  8   injected cells potentially important therapy

 

  9   parameters?

 

 10             For example, will injection of cells near

 

 11   the more ischemic endocardial surface of the heart

 

 12   provide therapy that is identical or equivalent to

 

 13   injection near the less ischemic epicardial

 

 14   surface, or is a minimally dispersed bolus of cells

 

 15   at each injection site equivalent to wider

 

 16   dispersion of cells at each injection site?

 

 17             We currently suspect that catheter design,

 

 18   cell suspension characteristics, and injection

 

 19   speed can all affect injection depth and spread.

 

 20             If a clinical study is performed using a

 

 21   specific injection catheter and a specific cell

 

 22   suspension, will the same therapy then be delivered

 

 23   if a different injection catheter is used to

 

 24   deliver that same cell therapy?

 

 25             Of note, you can use animal studies, and

 

                                                               306

 

  1   this is where the large animals become important,

 

  2   to characterize the depth and spread of the cell

 

  3   suspensions produced using a specific catheter.

 

  4             Finally, and this is a question I think

 

  5   that unites all the questions listed above on this

 

  6   slide, when an investigational therapy is studied,

 

  7   how important is it that the therapy delivered be

 

  8   characterized?  When an investigational therapy is

 

  9   poorly understood, is the characterization of

 

 10   therapy more or less important?

 

 11             Today's meeting is focused on scientific

 

 12   discussion of cell therapies for cardiac disease,

 

 13   and because of both the focus of this meeting and

 

 14   the time constraints, this is not a good forum for

 

 15   discussion of regulatory concerns related to

 

 16   cardiac catheters, however, if you would like to

 

 17   discuss cardiac catheters intended for delivery of

 

 18   cell therapies, you may contact either myself or my

 

 19   branch chief, Mr. Elias Mallis.

 

 20             I have listed our contact information on

 

 21   this slide.  Again, it will be posted on the web

 

 22   site.

 

 23             Finally, because this is one of the final

 

 24   presentations today, I have one final slide.  My

 

 25   manager has repeatedly asked the following

 

                                                               307

 

  1   question:  Whether there are earlier or predicate

 

  2   devices that have been used to provide catheter

 

  3   delivery of biologicals.

 

  4             He has also asked what we can learn from

 

  5   any earlier devices.  Initially, I was unable to

 

  6   define a useful predicate for catheter delivery of

 

  7   biologicals.  Then, I did find a useful example,

 

  8   and it was among photos from the Minnesota State

 

  9   Fair.

 

 10             The obvious lesson from this photo, I

 

 11   would say is that intense concentration may be

 

 12   required during catheter manipulation.

 

 13             [Laughter.]

 

 14             DR. JENSEN:  Thank you.

 

 15             [Applause.]

 

 16             DR. RAO:  Thank you, Dr. Jensen.

 

 17             I am going to suggest that we wait and

 

 18   hold off questions until we hear from Dr. Lederman,

 

 19   as well, since he may be perhaps answering some of

 

 20   those questions, and then direct questions to both

 

 21   people at the end of that talk.

 

 22        Transcatheter Myocardial Cell Delivery: Questions

 

 23               and Considerations from the Trenches

 

 24             DR. LEDERMAN:  I am going to be quick.  I

 

 25   am grateful for the opportunity to speak before

 

                                                               308

 

  1   this audience and this committee.  Thank you for

 

  2   your service.

 

  3             I am speaking to you as a clinical

 

  4   cardiologist, so I like to think of myself as Joe

 

  5   six pack of clinicians. I will be talking about the

 

  6   considerations, the frustration that many

 

  7   investigators feel when we would like to talk with

 

  8   the agency to get some guidance about how to start

 

  9   bringing these interesting therapeutics to clinic

 

 10   assuming we have determined that the timing is

 

 11   right to bring therapeutics to clinic.

 

 12             I am sorry there will be a bit of

 

 13   repetition.  I will try to go quickly through

 

 14   repetitive slides.

 

 15              We are dealing with integrated therapies

 

 16   and unfortunately, we are also dealing with a

 

 17   morass of regulatory purviews that don't

 

 18   necessarily intersect.  You have seen already that

 

 19   we have considerations of delivery devices, as well

 

 20   as cellular agents, to combine in therapy, and we

 

 21   haven't discussed much in this room combinations of

 

 22   novel mobilization agents should we choose to use

 

 23   that route to drive our cells, and it becomes

 

 24   difficult when we have proof of concept in some

 

 25   animal models to find an appropriate proof of

 

                                                               309

 

  1   concept to support our clinical trial and to

 

  2   support our safety considerations when some of our

 

  3   colleagues outside the U.S. have kind of moved

 

  4   ahead.

 

  5             So, let's see if I can generate some

 

  6   interesting questions for the committee, and that

 

  7   is really what I hope to end on.

 

  8             You have seen this slide from Strauer.  I

 

  9   will review again just the different approaches to

 

 10   cell delivery, into coronary cell delivery is

 

 11   attractive because it is very easy, there is a wide

 

 12   dispersion into the target territory, and there are

 

 13   a lot of available devices to be used although they

 

 14   must be used off label.

 

 15             The disadvantages have been mentioned that

 

 16   there is a potential coronary artery injury.  One

 

 17   of the real clever innovations by Strauer's group

 

 18   and by Sawyer's group and the Hanover group is that

 

 19   they have chosen to deliver cells through an

 

 20   occlusive balloon deployed at the site of a

 

 21   recently deployed stent, so there is really almost

 

 22   no possibility of coronary injury from the delivery

 

 23   device, however, there is a possibility of a

 

 24   coronary microembolism, and when you test this is

 

 25   the setting of a recent acute myocardial

 

                                                               310

 

  1   infarction, that coronary embolism may be difficult

 

  2   to detect, so clinicians have gotten away with it,

 

  3   or at least I should say their patients have gotten

 

  4   away with it.

 

  5             Certainly, there is a great potential for

 

  6   direct washout of injected cells.  Very few people

 

  7   have actually measured this or reported this, but

 

  8   there is some evidence that there is a low

 

  9   fractional retention of delivered cells, and really

 

 10   this intracoronary cell delivery is yet another way

 

 11   to expose the entire patient to the therapeutic

 

 12   agent.

 

 13             This kind of approach is probably

 

 14   unsuitable to certain patient populations,

 

 15   especially those with chronic myocardial ischemia

 

 16   when the inflow arteries are occluded. Most

 

 17   investigators have taken advantage of transient

 

 18   coronary flow interruption ostensibly to improve

 

 19   local retention, but it is just not clear the value

 

 20   or importance of this transient coronary flow

 

 21   interruption.  Certainly, a lot of patients can't

 

 22   tolerate prolonged coronary flow interruption

 

 23   without incremental myocardial injury.

 

 24             There has been discussion about both

 

 25   surgical and transcatheter cell injection. 

 

                                                               311

 

  1   Certainly, most of us will recognize that

 

  2   catheter-based injection is less morbid than

 

  3   surgical epicardial injection.  Primary surgery has

 

  4   been unattractive in investigational studies when

 

  5   the surgery is offered only for the sake of cell

 

  6   delivery.

 

  7             The problem is in small studies, combining

 

  8   cell delivery with an effective therapy, I think

 

  9   has been mentioned by several people before,

 

 10   doesn't really generate meaningful safety or

 

 11   efficacy data, because the assessment of toxicity

 

 12   events is confounded by the concomitant surgical

 

 13   procedure, and the assessment of efficacy events is

 

 14   very easy to ascribe to the concomitant effect of

 

 15   therapy.

 

 16             So, I think this kind of approach should

 

 17   probably be discouraged in small, single-center

 

 18   studies.  I hope some investigators in the room

 

 19   have already overcome this in moving to larger

 

 20   studies.

 

 21               Direct catheter injection of the

 

 22   myocardium is attractive because we can achieve a

 

 23   high local cell density and probably a high total

 

 24   dose.  It is certainly very easy also, with the

 

 25   variety of catheters that I will describe, and the

 

                                                               312

 

  1   entire myocardium is for the most part accessible

 

  2   irrespective of the patient's individual coronary

 

  3   anatomy.

 

  4             But these devices are disadvantageous in

 

  5   the U.S. because there are no approved devices,

 

  6   although a few are available through

 

  7   investigational device exemption.

 

  8             I will show you some data that there also

 

  9   is low retention of injected cells and that direct

 

 10   myocardial injection is yet another means of

 

 11   systemic exposure of the patient to the cellular

 

 12   agent.  We are left even in the best situation with

 

 13   multifocal cell accumulation, meaning a

 

 14   heterogeneous dispersion of the cellular agent.

 

 15             Whether or not that is important isn't

 

 16   clear, and there is the potential for damage to the

 

 17   myocardium or to the chordal structures or the

 

 18   valve structures, however, this potential has not

 

 19   been supported by experience with comparably

 

 20   aggressive or more aggressive intramyocardial

 

 21   catheters especially in the fairly large laser

 

 22   myocardial "revascularization" experience or

 

 23   angiogenic gene transfer.

 

 24             There are a bunch of variants of

 

 25   myocardial injection catheters.  There are

 

                                                               313

 

  1   techniques to access the pericardium and bathe the

 

  2   epicardial surfaces of the heart with the cell

 

  3   preparation of interest.

 

  4             Patients with chronic myocardial ischemia

 

  5   who have undergone coronary bypass surgery are, for

 

  6   the most part, not eligible, so this is a difficult

 

  7   approach in early clinical trials.

 

  8             There are investigators who have

 

  9   demonstrated satisfactory delivery of genes and

 

 10   cells by retrograde coronary venous approach.  I

 

 11   will show you a picture to show what that means.  A

 

 12   company has commercialized tangential transvenous

 

 13   intramyocardial injection.

 

 14             Then, there are a bunch of endocavitary

 

 15   catheters that go across the aortic valve

 

 16   retrograde and are pretty successful in delivering

 

 17   cellular agents.  There are an array of what I call

 

 18   "dumb" catheters that we use just under x-ray

 

 19   guidance that are very attractive because they are

 

 20   quick and easy.  Two examples are Boston Scientific

 

 21   Stilletto and Biocardia Device.

 

 22             There a couple of smarter devices, it is

 

 23   not clear that they are better, but they employ a

 

 24   Static Roadmap like the Cordis Biosense

 

 25   electromagnetic guidance system with some

 

                                                               314

 

  1   non-fluoroscopic guidance.  There are also devices

 

  2   that have an integrated ultrasound, and the

 

  3   smartest devices, I will show you an example, in

 

  4   research mode only, of instantaneous imaging both

 

  5   of the tissue and the device.

 

  6             We should all open our hearts to the

 

  7   possibility that surgical videothoracoscopic, a

 

  8   minimally invasive surgical procedure may

 

  9   accomplish cell delivery with very little morbidity

 

 10   even in a primary surgical procedure.

 

 11             This is just a demonstration of one of the

 

 12   so-called "dumb" catheters.  This is a Boston

 

 13   Scientific catheter going from a femoral artery of

 

 14   a pig, across the aortic valve, and can

 

 15   successfully guide whatever agent you want with

 

 16   centimeter, not millimeter, precision to any aspect

 

 17   of the endocardial surface.

 

 18             Medtronic has recently bought

 

 19   Transvascular, which is an interesting device that

 

 20   has an integrated ultrasound to guide the

 

 21   deployment of a needle through a coronary vein and

 

 22   can access target myocardium through any of the

 

 23   coronary veins in a tangential fashion.  This has

 

 24   been tested in clinic in a very small number of

 

 25   patients, and so far there haven't been any safety

 

                                                               315

 

  1   disasters.

 

  2             A retrograde transvenous approach has been

 

  3   demonstrated I believe only in animals.  The

 

  4   Stanford group, Keith Marsh's group, have been

 

  5   interested in this for delivery of dyes or gene

 

  6   agents or even some cellular agents.  I am sorry

 

  7   the pictures aren't very attractive.  It is not

 

  8   clear to me how this can possibly work, but the

 

  9   proof of principle has been shown.

 

 10             It is also attractive in that the inflow

 

 11   coronary artery anatomy is not a problem since

 

 12   coronary vein patency is maintained in virtually

 

 13   all patients.  Dr. Perin is an expert in the use of

 

 14   the Cordis electromagnetic guidance system,  the

 

 15   Biosense system that has its advantages in that it

 

 16   has been widely used and tested for a variety of

 

 17   investigational approaches.

 

 18             It is disadvantageous in that it is a

 

 19   prior roadmap of the heart that may vary over time,

 

 20   and so it is not clear to me that you accomplish

 

 21   millimeter scale precision of your injections, but

 

 22   it is also not clear that that is very important.

 

 23             I think Dr. Epstein's group also has great

 

 24   expertise in the use of the device for cell

 

 25   therapy.

 

                                                               316

 

  1             Just to brag about some work in my lab for

 

  2   a moment, we have used real-time MRI to guide cell

 

  3   injection to very small targets with great

 

  4   precision and great ease. This is, as you see, a

 

  5   multi-slice real-time movie of the heart.  You can

 

  6   see it in long axis and short axis, of a pig in

 

  7   which we have caused a tiny, little

 

  8   microinfarction, shown in white.

 

  9             The catheter is shown in red and green,

 

 10   and we can see with great 3-dimensional sense where

 

 11   we are steering our catheter, and if we like, we

 

 12   can label cells, say, mesenchymal stromal cells,

 

 13   very easily.

 

 14             Here, the mesenchymal stromal cells are

 

 15   showing up in black, so you can actually see

 

 16   interactively, as you deliver the cells, that at

 

 17   least some are attained in the target myocardium,

 

 18   and this is almost ready for clinical application.

 

 19             I want to show a little bit of data.  This

 

 20   is from my youth when I was just out of fellowship,

 

 21   a study that was funded by Boston Scientific while

 

 22   I was at still at University of Michigan.

 

 23             We injected neutron-activated microspheres

 

 24   into the heart with an endocavitary catheter,

 

 25   direct surgical approach or postmortem, and what

 

                                                               317

 

  1   was interesting is that we inject and then kill the

 

  2   animal within minutes, and most of what we inject,

 

  3   both surgically or transcatheter approach, is lost

 

  4   immediately.

 

  5             There is some effect of volume.  Smaller

 

  6   volumes were associated with slightly greater

 

  7   retention in tissue. This is 10 microliters.  That

 

  8   is a tiny, tiny injection compared with 20 or 100.

 

  9             A better study was published by Smits from

 

 10   the Thoraxcenter group in Rotterdam using

 

 11   scintigraphy and radiolabeled albumin, either plain

 

 12   radiolabeled albumin or a colloid, and they showed

 

 13   also loss of the majority of injectate after just a

 

 14   minute by scintigraphy.

 

 15             Their colloidal preparation had great

 

 16   retention,  which is interesting, and that there

 

 17   might, of course, be some interaction between

 

 18   biological agents and the myocardial interstitium,

 

 19   so conceivably, cells won't necessarily be lost.

 

 20   These are studies that can be done in the lab in

 

 21   healthy animals.

 

 22             But I think it is easy to say that local

 

 23   myocardial injection is at best an exaggeration,

 

 24   that most injectate is lost rapidly and exits

 

 25   either by backflow directly into the myocardial

 

                                                               318

 

  1   cavity, which we can see directly, or with

 

  2   intracardiac ultrasound or even with

 

  3   high-resolution x-ray, that there is also a clear,

 

  4   what I call "intravasation" or return to the

 

  5   coronary circulation or coronary lymphatics.

 

  6             When we inject too deeply and directly

 

  7   into the pericardium, that is another mode of exit,

 

  8   but clearly, the interstitial myocardial target

 

  9   retains only a fraction of what we intend to

 

 10   deliver there.

 

 11             So, where does this material go and is

 

 12   that really important?  I think it is important at

 

 13   least that we assume that what we think we are

 

 14   injecting by any route goes everywhere, and I think

 

 15   that means that conventional toxicology or

 

 16   biodistribution experiments can be conducted in

 

 17   uninfarcted animals without the needle of interest

 

 18   just by modeling it as a left atrial or left

 

 19   ventricular cavitary injection that is not device

 

 20   specific.

 

 21             I think also it is interesting to talk

 

 22   about open- label autologous unfractionated bone

 

 23   marrow data, what is the incremental value of

 

 24   animal safety or tox experiments in light of the

 

 25   fact that provisional safety has been shown in

 

                                                               319

 

  1   open-label studies if we are convinced that the

 

  2   safety reporting has been complete, and I am pretty

 

  3   comfortable that it has been.

 

  4             For autologous leukapheresis products, say

 

  5   we apherese CD34 cells for a direct myocardial

 

  6   injection, those cells are circulating already.  It

 

  7   is not clear to me what is the value of incremental

 

  8   biodistribution experiments regarding systemic

 

  9   exposure.

 

 10             I will qualify that by saying that

 

 11   allogeneic material perhaps should be treated

 

 12   differently, but for autologous material, it is not

 

 13   clear to me that these animal data are valuable,

 

 14   and to require it of investigators before going to

 

 15   clinic sounds dubious in my opinion.

 

 16              Also, it is worth noting that the

 

 17   importance of targeting is just not established.

 

 18   While I am very interested in precise anatomic

 

 19   targeting, it is not clear why we need it.

 

 20             Delivery targets certainly vary by

 

 21   application.  We may want to target infarct

 

 22   borders.  Doris Taylor had some data today that

 

 23   infarct borders may be unattractive for certain

 

 24   therapies.

 

 25             Do we want to target ischemic zones, or do

 

                                                               320

 

  1   we want to avoid ischemic zones, do we want to

 

  2   avoid thin myocardium?  Is roadmap data worse than

 

  3   blind data or worse than instantaneous real-time

 

  4   MRI data?  It is just not clear.

 

  5             Certainly, good targeting is attractive in

 

  6   that it may reduce overlapping injections and waste

 

  7   of injections, and overlapping injections may

 

  8   increase systemic loss.  It is hard to imagine that

 

  9   we might be exceeding some therapeutic index.

 

 10             So, in other words, the value of targeting

 

 11   is just not clear to me, and if we are able to show

 

 12   some efficacy, it is not clear how much we must ask

 

 13   of investigators to establish these catheter-based

 

 14   information before going to clinic.

 

 15             A point that has not been mentioned, but

 

 16   that I have encountered in animal studies is that

 

 17   operators need feedback regarding delivery of their

 

 18   therapeutic agent, and I would like to encourage

 

 19   the committee and the regulatory agency to consider

 

 20   contrast labeling at the time of cell delivery.  It

 

 21   is certainly clearly more important than needle

 

 22   stability measures.

 

 23             There are lots of ways to label injection

 

 24   mixtures.  You can admix contrast into your

 

 25   injection cocktail.  Iodinated radiocontrast is

 

                                                               321

 

  1   clearly tolerated in myocardium.  We inject

 

  2   high-dose, full-strength intracoronary

 

  3   radiocontrast, replacing blood inflow for many

 

  4   seconds in patients in all settings, acute MI,

 

  5   acute and chronic ischemia, and healthy myocardium.

 

  6             It is certainly very well tolerated.  It

 

  7   is hard to believe that iodinated radiocontrast

 

  8   injected into the myocardial interstitium is not

 

  9   tolerable.  It is certainly well tolerated in

 

 10   animal experiments, but this kind of feedback under

 

 11   x-ray guidance, for example, is critical in knowing

 

 12   that we are delivering the cells into the target

 

 13   that we think we are.

 

 14             If we are doing injections under MRI,

 

 15   then, certainly we can admix gadolinium-based MRI

 

 16   contrast agents in very dilute form just like the

 

 17   agent that reaches the myocardium after systemic

 

 18   exposure, and these are very easy to be tested

 

 19   biocompatible in vitro.

 

 20             Certainly, we can do test injections of

 

 21   contrast to test the purchase of our needle in the

 

 22   myocardium before injecting the cell of interest,

 

 23   but the problem with these test injections of

 

 24   radiocontrast is that the catheter and hub dead

 

 25   space often exceeds the volume of the desired cell

 

                                                               322

 

  1   injection, so that is a problem that is difficult

 

  2   to overcome.

 

  3             Alternatively, we can label our cells.

 

  4   That is certainly easy if we want to deliver our

 

  5   cells under MR or under echo, but it is not clear

 

  6   what options we have under x-ray, so to the members

 

  7   of this committee, please facilitate solutions to

 

  8   this clinical need.

 

  9             Some engineering concerns have been

 

 10   mentioned in the very excellent guidance materials

 

 11   supplied to members of the committee.  I just want

 

 12   to speak to some of them.

 

 13             The issues of biocompatibility of lumens

 

 14   and potential clogging of lumens, this is easy to

 

 15   test on benchtop and doesn't require animal

 

 16   experiments.

 

 17             The issues of balloon injury of target

 

 18   coronary arteries is an important one.  You have

 

 19   seen creative solutions by investigators in Europe

 

 20   by protecting the target coronary artery, inflating

 

 21   their occlusion balloon inside a recently deployed

 

 22   stent.

 

 23             Certainly, there are noninjurious

 

 24   compliant occlusion balloons that are clinically

 

 25   approved for a variety of peripheral artery

 

                                                               323

 

  1   applications, that are used widely in the cerebral

 

  2   circulation, that are also used for coronary

 

  3   protection and substantial equivalence data are

 

  4   already widely available.

 

  5             We have also heard discussion of

 

  6   considerations of the pressure capacity of balloon

 

  7   wire lumens.  The European investigators, for

 

  8   example, are administering their cells via the wire

 

  9   lumen of an inflated coronary balloon.

 

 10             As a practicing interventional

 

 11   cardiologist, I don't consider that an important

 

 12   concern because every balloon, every over-the-wire

 

 13   design balloon that I put into a patient, I expect

 

 14   to use for intracoronary angiography, and I use for

 

 15   intracoronary angiography with a fairly high

 

 16   pressure system.

 

 17             There are many times I need to know that

 

 18   my balloon is in the right place, so I pull out the

 

 19   wire and I inject contrast at a fairly high

 

 20   pressure directly through the balloon lumen.

 

 21             That is not, of course, an indicated use,

 

 22   but it is a wide use by all operators of coronary

 

 23   artery balloons, and I think the test of time has

 

 24   already been past, but if you like simple benchtop

 

 25   pressure data, they are easy to acquire.

 

                                                               324

 

  1             Regarding endomyocardial injection

 

  2   catheter engineering concerns, the same

 

  3   biocompatibility and clogging concerns are easy to

 

  4   answer on benchtop tests.  This issue of variable

 

  5   needle extension is probably an important one if

 

  6   injection depth proves to be important, and it is

 

  7   not clear that it is, but this can be addressed in

 

  8   benchtop testing.

 

  9             It has been mentioned that purchase

 

 10   stability is important to assure injectate reaching

 

 11   the target tissue.  My assertion about marking or

 

 12   labeling injection cocktails with contrast might

 

 13   address that concern.

 

 14             The report from UC/SF from Jonathan

 

 15   Coleman using an old intracardiac echo device

 

 16   reporting instability is actually a spurious

 

 17   observation because of through-plane motion of the

 

 18   target that the UC/SF group was inspecting with a

 

 19   fixed vena caval or a right atrial intracardiac

 

 20   echo device.  In other words, I don't think it has

 

 21   been shown that the contact of EP catheters or

 

 22   myocardial injection catheters cannot be

 

 23   maintained, in fact, just the opposite, especially

 

 24   from the Biosense device which has a local cardiac

 

 25   electrogram capability to assure contact stability.

 

                                                               325

 

  1             So, I think this is not really an

 

  2   important problem for us to worry about.

 

  3             The issue of potential myocardial

 

  4   perforation is often raised when we discuss the

 

  5   possibility of delivering cells directly into

 

  6   patients after a recent large myocardial

 

  7   infarction, and I think that is an important one.

 

  8   It is interesting that you refer to myocardial

 

  9   biopsy devices as predicate devices, because as I

 

 10   view the biotomes as some of the most dangerous

 

 11   devices we ever laid hands on, they are so

 

 12   incredibly stiff and indeed perforations do

 

 13   sometimes occur.

 

 14             Fortunately, there is an animal experience

 

 15   from my lab.  I guess we should probably get it out

 

 16   there, of a large number of injections directly

 

 17   into freshly infarcted myocardium, and I think this

 

 18   kind of data is easy to obtain.

 

 19             But the bigger issue is not that freshly

 

 20   infarcted myocardium can be safety injected, it is

 

 21   that the device companies can't really control the

 

 22   operators.  I have seen this so many times.  An

 

 23   engineer walks into a lab and cringes as the

 

 24   interventional cardiologist effectively abuses the

 

 25   device.  How do you model operator misbehavior? It

 

                                                               326

 

  1   is kind of difficult.

 

  2             In reality, proof of principle has been

 

  3   established.

 

  4             The issue of inadvertent pericardial

 

  5   injection probably has little or no clinical

 

  6   importance especially when compared with the loss

 

  7   of injectate via other routes, and its only value

 

  8   is that you are not delivering what you think

 

  9   directly into target tissue again, the value of

 

 10   instantaneous visualization of injections.

 

 11             The issue of distribution of injected

 

 12   material within the target myocardium, I think it

 

 13   may be reasonable to assume that this distribution

 

 14   is different in normal myocardium versus fresh

 

 15   infarct versus chronic scar, but the value of these

 

 16   data are just not clear compared with the efficacy

 

 17   data in support of preclinical or early clinical

 

 18   experiments, so having this information of how many

 

 19   cubic centimeters of myocardium are exposed to

 

 20   target cell based on a given volume or dose of

 

 21   cells, it is just not clear why we need that

 

 22   information.  This kind of information ultimately

 

 23   can only be valuable in patients.

 

 24             Are endomyocardial injection catheters

 

 25   generic?  In my opinion, assuming benchtop

 

                                                               327

 

  1   biocompatibility has been determined, and assuming

 

  2   that mechanical performances are satisfactory

 

  3   compared with predicate devices, I think that a

 

  4   myocardial injection catheter is pretty much the

 

  5   same from one to another.

 

  6             One needle device should be translatable

 

  7   to another, and the scientific and regulatory value

 

  8   of additional data from large mammals, healthy

 

  9   ischemic infarcted, is really pretty small and hard

 

 10   to justify, in my opinion.  So, I keep giving this

 

 11   message, nihilistic message about the large animal

 

 12   models investigators have been asked to provide.

 

 13             To summarize my opinions, I think

 

 14   engineering and biocompatibility concerns can be

 

 15   addressed with benchtop data.  I think that animal

 

 16   model safety experiments matching a given catheter

 

 17   device with a given putative therapeutic agent

 

 18   don't meaningfully contribute to patient safety and

 

 19   are, in fact, potentially misleading.

 

 20             I wish that there were a way to get a

 

 21   screening IDE or IND capability to support testing

 

 22   new cell preparations without repeating unnecessary

 

 23   preclinical experiments as we switch from device to

 

 24   device, and ultimately, careful human

 

 25   experimentation is what is most important.

 

                                                               328

 

  1             Let me just make a few more points before

 

  2   I turn to some questions and step off the podium,

 

  3   so people can go home.

 

  4             I want to reiterate some points made by

 

  5   other speakers today, and I want to reiterate it

 

  6   especially to the regulatory officers here.  I

 

  7   think that blinded placebo groups are mandatory

 

  8   even in first experiments.

 

  9             Why would you conduct an experiment

 

 10   without a suitable matched control in the name of

 

 11   safety?  That is just bad science.  Interestingly,

 

 12   there has not been a single open-label or "Phase I"

 

 13   safety trial that fails to make an efficacy claim

 

 14   without a suitable matched control.

 

 15             Unfortunately, the agency is inadvertently

 

 16   discouraging blinded controls, for example, when

 

 17   they ask for a delay in between exposing a given

 

 18   subject within a given group of patients, asking

 

 19   for seven days or four-week delay between patients

 

 20   to look for safety of individual patients.  This

 

 21   often frightens investigators away and makes them

 

 22   drop placebo groups.

 

 23             I think that in cardiology, we rarely

 

 24   conduct classic Phase I studies in end-stage

 

 25   subjects in spite of the conversation in Doris

 

                                                               329

 

  1   Taylor's speech.

 

  2             So, I would like to encourage people in

 

  3   the committee and encourage regulatory agencies to

 

  4   facilitate inclusion of blinded placebos in early,

 

  5   first in man even, experiments.

 

  6             I was also asked to talk a little bit

 

  7   about the safety of direct myocardial injection,

 

  8   and unfortunately, there are no large series of

 

  9   direct myocardial injection of cells or any other

 

 10   agents, however, a related catheter has been tested

 

 11   in a few hundred patients.

 

 12             Cordis had a myocardial--they called it a

 

 13   DMR, direct myocardial revascularization procedure,

 

 14   but it was a way to burn the myocardium from a

 

 15   transcatheter approach.  In this Cordis-sponsored

 

 16   study presented by Martin Leon and Ron Kornowski a

 

 17   few years ago, a Cordis Biosense derivative,  a

 

 18   device much like the myocardial injection catheter

 

 19   shown today, was used to steer into the myocardium

 

 20   of 300 patients with refractory ischemia, mild or

 

 21   moderate left ventricular dysfunction, and preserve

 

 22   wall thickness.

 

 23             One hundred patients underwent sham

 

 24   procedures, placebo burns of the myocardium, and

 

 25   200 more received laser in two different doses. 

 

                                                               330

 

  1   This is just the clinical complications.  I will

 

  2   point you to left ventricular perforation.  There

 

  3   were none in the 100 placebo patients, there was 1

 

  4   out of 100 in the highest laser dose, which is

 

  5   comparable to some other laser trials.

 

  6             I think this establishes a relative safety

 

  7   base from a perforation perspective.  These other

 

  8   events unfortunately weren't very well described,

 

  9   and this study unfortunately has not been

 

 10   published, and I am not sure it ever will be

 

 11   submitted for publication, but the acute safety of

 

 12   this device, I think is relatively self-evident.

 

 13             So, is placebo and the myocardial

 

 14   injection safe in principle?  I think yes, and it

 

 15   is not a reason to discourage these placebo groups

 

 16   in early first clinical studies.

 

 17             So, again, we are trying to bring

 

 18   therapies to clinical testing, and we are trapped

 

 19   between delivery devices and cellular agents that

 

 20   we would like to use together.

 

 21             I want to just ask a few hypothetical case

 

 22   questions to the committee before I step down.  I

 

 23   certainly don't have answers, but I hope you find

 

 24   it provocative.

 

 25             Let us say that an investigator identifies

 

                                                               331

 

  1   a novel marker, an HL321, let's call it, that

 

  2   identifies some kind of progenitor cell for which

 

  3   there is no clear animal homolog.

 

  4             There are some limited preclinical

 

  5   efficacy that when an enriched human HL321

 

  6   population is injected to intrinsically

 

  7   immunocompromised rat infarct, it causes functional

 

  8   recovery compared with a population of known cells

 

  9   that are relatively depleted, so this is not the

 

 10   most robust type of experiment, but based on

 

 11   experiments like this, and based on patients who

 

 12   are clamoring for therapy of their massive

 

 13   myocardial infarction, investigators may want to

 

 14   bring some test of this therapy to clinic even now.

 

 15             How could we test local autologous HL321

 

 16   cells assuming we had a feasible way to mobilize

 

 17   and recover these cells?  How could we test that

 

 18   for safety and efficacy?

 

 19             In the example I want to give you, there

 

 20   already is a commercial cell system available,

 

 21   marked with a CE in Europe, and in Europe, hundreds

 

 22   of patients have successfully undergone bone marrow

 

 23   transplantation with a population positive

 

 24   selection for this marker, and also in Europe,

 

 25   dozens of patient underwent local cardiac delivery

 

                                                               332

 

  1   trials in a variety of applications, and Phase II

 

  2   trials have been offered.

 

  3             Is it unreasonable to permit U.S.

 

  4   investigators to conduct similar experiments now

 

  5   that provisional safety has been tested in Europe,

 

  6   if incompletely reported in Europe?

 

  7             For this kind of clinical experiment, are

 

  8   additional animal data really necessary when human

 

  9   studies have already been conducted, and when there

 

 10   is no animal homolog of that positive selection

 

 11   marker?  What animal model is really adequate?

 

 12             Can we use the experience of investigators

 

 13   like Dr. Epstein, investigators like Dr. Perin,

 

 14   using undifferentiated bone marrow to support the

 

 15   local delivery of other autologous cells, and are

 

 16   individual cell preps, autologous cell preps

 

 17   substantially equivalent in this case when they

 

 18   have just been derived directly from the patient

 

 19   irrespective of sources?  Is bone marrow that much

 

 20   different from apheresis product, from a

 

 21   mobilization product?

 

 22             So, how can we apply non-U.S.A. human

 

 23   safety data to support U.S. clinical trial

 

 24   proposals, or should we continue the way we are now

 

 25   and just sit and wait for others to do their

 

                                                               333

 

  1   experiments without us?

 

  2             Thank you very much for your attention,

 

  3   and I hope you found these questions provocative.

 

  4             [Applause.]

 

  5             DR. RAO:  We have time for a few

 

  6   questions.

 

  7                               Q&A

 

  8             DR. NOGUCHI:  I think that was a terrific

 

  9   representation of the tension between belief and

 

 10   what is published or not published, and what we

 

 11   might call the paradigm for FDA, which is absence

 

 12   of evidence is not evidence of absence.

 

 13             I will just challenge you a little bit.

 

 14   When we   see safety data whether it is from

 

 15   another country or here, one of the critical

 

 16   questions for an adverse event is, well, is that

 

 17   really showing that if you give a product that you

 

 18   have a lack of an adverse event, or could it be

 

 19   that, in fact, you didn't give a product, which by

 

 20   your own arguments you would say most of the time,

 

 21   you lose most of the product all over the place.

 

 22             So, some of it falls into the category of

 

 23   our experience is that adverse events actually

 

 24   other than from the actual injections of others for

 

 25   biological products don't occur unless you actually

 

                                                               334

 

  1   also have bioactivity, and sometimes if you don't

 

  2   have any cells, you may not have any bioactivity.

 

  3             So, I think that you have a number of very

 

  4   genuine points very well worth arguing, but I would

 

  5   just caution that it is very simple to say

 

  6   something is safe.  We rarely say look at all the

 

  7   published data and it's safe, because we don't

 

  8   really know if a product was actually being used

 

  9   there, and perhaps that is one of the points you

 

 10   might want to just think about.

 

 11             DR. LEDERMAN:  I, of course, can't even

 

 12   answer your question, and I want to point out I

 

 13   want to thank the regulatory officers and the FDA

 

 14   for trying to protect our patients, and to try to

 

 15   protect the American public.  You are in a very

 

 16   difficult position that is often a thankless

 

 17   position, but I just hope you are open to this kind

 

 18   of conversation.

 

 19             DR. NOGUCHI:  Absolutely.  That is why we

 

 20   would like to have you end it here, because I am

 

 21   sure it will provide the focus of discussion for

 

 22   tomorrow.

 

 23             DR. RAO:  Any other questions?

 

 24             DR. EPSTEIN:  Bob, I really enjoyed that

 

 25   presentation, it was really great.  I would just

 

                                                               335

 

  1   like to raise one point.  For example, to my

 

  2   knowledge, no one has really tested in depth the

 

  3   safety of the transvascular administration of an

 

  4   angiogenic agent or its cells.

 

  5             The reason I feel that might be different,

 

  6   for example, than a transendocardial or a

 

  7   transepicardial injection is because you are

 

  8   injecting it right around a large artery.  It is

 

  9   conceivable that there can be pro-atherosclerotic

 

 10   or pro-restenotic effects.

 

 11             DR. LEDERMAN:  I am sorry.  Let me

 

 12   interrupt your question.  Do you mean intracoronary

 

 13   approach or the Medtronic transvascular approach?

 

 14             DR. EPSTEIN:  The Medtronic approach, so

 

 15   where you are injecting it, not downstream at the

 

 16   small vessel level, but at the large vessel level,

 

 17   so if you are injecting it, for example, through

 

 18   the venous system, it is contiguous with the

 

 19   arteries that have atherosclerosis in it.

 

 20             So, I would think that for that special

 

 21   case, the FDA would require that you have to show

 

 22   that there is no deleterious effect in terms of a

 

 23   pro-atherosclerotic effect. I would be interested

 

 24   in your thoughts about that.

 

 25             DR. LEDERMAN:  I think that point is well

 

                                                               336

 

  1   taken and every such safety request or demand is

 

  2   interesting and valuable, but how do we answer that

 

  3   kind of question satisfactorily.

 

  4             Let's take the question you just asked, a

 

  5   tangential myocardial needle or perhaps a

 

  6   retrograde venous administration of agent X, and

 

  7   the problem of an unrecognized atherogenic effect,

 

  8   how on earth do we test that?  Are apoE knockout

 

  9   mouse experiments satisfactory?

 

 10             DR. EPSTEIN:  For this particular, I would

 

 11   injure a vessel in a pig, and then inject whatever

 

 12   agent you are interested in transvenously in the

 

 13   area of that injured vessel and just see whether

 

 14   there is an increase in the neointimal response.

 

 15             I don't know how you carry--I mean your

 

 16   question would be so what, whatever you see, and

 

 17   that would be a good question.

 

 18             DR. LEDERMAN:  But that is exactly right,

 

 19   that might reassure us, but that is also not

 

 20   atherosclerosis.

 

 21             DR. EPSTEIN:  That's right, but the AMI

 

 22   studies, you know, you are doing angioplasty, so if

 

 23   you were to increase the incidence of restenosis in

 

 24   the pig, you know, quite predictively, it would

 

 25   certain add a major cautionary note to approval of

 

                                                               337

 

  1   such a protocol.

 

  2             DR. TAYLOR:  I would ask two questions.

 

  3   One, very short, but one is you said that you would

 

  4   argue that exogenous delivery of any given cell

 

  5   population is equivalent essentially, but I would

 

  6   think that the GCSF, or if you have given one bone

 

  7   marrow cell population, is it right to go ahead

 

  8   with all the others without necessarily more safety

 

  9   data?

 

 10             I would argue that the GCSF data that just

 

 11   came out would actually argue the converse, that

 

 12   the only difference there was mobilization of cells

 

 13   that would otherwise be endogenous to that same

 

 14   patient, and yet an increased number of those cells

 

 15   clearly caused some negative effect.

 

 16             DR. LEDERMAN:  I wasn't actually making

 

 17   that assertion.  I was making the assertion that

 

 18   needle injection catheters are ultimately very

 

 19   similar.

 

 20             DR. KURTZBERG:  But the difference, there

 

 21   have been, I don't know how many tens of thousands

 

 22   of patients have had GCSF, we have gotten bone

 

 23   marrow in their right atrium, so I mean the

 

 24   dissemination of those cells is not the issue, it

 

 25   is combining that with a local technical injection

 

                                                               338

 

  1   and trauma to that site that is different.

 

  2             I mean there is experience with these

 

  3   cells disseminated through the human body for two

 

  4   decades, so that is not the issue.  The issue is

 

  5   what do the cells do in the setting of a local

 

  6   technical injection into an artery or other part of

 

  7   the heart that is sick.

 

  8             DR. TAYLOR:  I think that is sort of the

 

  9   point I am trying to make, that exogenous delivery

 

 10   is not necessarily the same thing as mobilization

 

 11   of cells, and that having more cells there, that we

 

 12   don't understand, we can't just interpolate from

 

 13   other data.

 

 14             I want to ask a very short question.  What

 

 15   do you think about clinicians moving forward who

 

 16   don't have experience with preclinical studies?  I

 

 17   mean one of the things that probably enabled

 

 18   Philippe to do the studies he did is he had that

 

 19   six years of preclinical experience making mistakes

 

 20   or whatever.

 

 21             What do you think about any clinician

 

 22   moving forward in a trial without having previous

 

 23   preclinical experience?

 

 24             DR. LEDERMAN:  My short answer is who

 

 25   cares what I think, and we all operate as parts of

 

                                                               339

 

  1   teams with expertise in our respective areas.  In

 

  2   cell therapy, for example, it would be outrageous

 

  3   for me to do an early clinical study without the

 

  4   close collaboration of cell therapy experts like

 

  5   some that are fortunately in the room.

 

  6             And would we need to have our local

 

  7   on-site preclinical experiments?  It is not clear

 

  8   to me how important that is.  It is more important

 

  9   to me that our agents be well characterized, that

 

 10   the studies be well conducted, and that they be

 

 11   designed in a way that the data can be interpreted

 

 12   rather than open-label, early clinical experiments

 

 13   that are very difficult to interpret.

 

 14             DR. RAO:  I really agree with on the

 

 15   emphasis you made about the fact that you should

 

 16   have a placebo-controlled trial, but then on the

 

 17   same token, you know, you also said that one

 

 18   catheter is much like the other, but I don't think

 

 19   that we can extrapolate that from saying one is

 

 20   much like the other, you know.

 

 21             I mean we worry about drugs when we say

 

 22   whether it is a generic formulation or whether it

 

 23   is a formulation which contains the same active

 

 24   ingredient, and to me, when you are looking at the

 

 25   device, you are making it with cells, you have to

 

                                                               340

 

  1   worry about how clearly or how similar the device

 

  2   is to any other device.

 

  3             We can't simply say, well, you know, the

 

  4   benchtop pressure was the same.  You know, I can

 

  5   take syringes and I can show you that the benchtop

 

  6   pressure on that cell agent injection is exactly

 

  7   the same because of how I do it, but, you know, I

 

  8   can put cells through it, and I can guarantee you

 

  9   that there would be a difference.

 

 10             I mean Dr. Menasch showed in his data you

 

 11   use a 27-gauge needle, and it is very different

 

 12   from using a 29-gauge needle.  It doesn't matter

 

 13   whether you have got the same pressure or not.

 

 14             So, I think that it would be hard put at

 

 15   least for me to be convinced that most catheters,

 

 16   even if they are giving delivery externally in much

 

 17   the same way, that one can logically extend it and

 

 18   say that it will probably be the same.

 

 19             DR. LEDERMAN:  So you are telling me--and

 

 20   I don't mean to belabor the point--but if you have

 

 21   two catheters by two different vendors, that have

 

 22   satisfactory benchtop testing for biocompatibility,

 

 23   have satisfactory hydraulic characteristics, that

 

 24   what you inject at one end comes out the other end,

 

 25   and one such catheter has satisfactory efficacy

 

                                                               341

 

  1   data in some kind of preclinical model, that you

 

  2   would require a repetition of that preclinical

 

  3   model for another catheter that is virtually the

 

  4   same?

 

  5             DR. RAO:  I am saying that right now we

 

  6   can't make the assumption that it will be the same,

 

  7   and the reason I say that is that we know that when

 

  8   we make minor manipulations to cells which we are

 

  9   delivering, for example,  if we take CD34 cells,

 

 10   which have been kept in culture for 48 hours as

 

 11   opposed to 12 hours, we have a very different

 

 12   endpoint result.  We know that.

 

 13             We don't know what the interaction will be

 

 14   with the catheter, and we can't make the assumption

 

 15   that because we know five parameters, that those

 

 16   will be adequate in making a reasonable prediction,

 

 17   so until I have a lot more data, I will be very

 

 18   surprised that one could make that statement or

 

 19   anybody would agree that that is okey.

 

 20             DR. LEDERMAN:  The end result is that we

 

 21   have an unmanageable number of permutations, an

 

 22   unmanageable number of permutations that makes it

 

 23   hard to make progress.

 

 24             DR. RAO:  But again I think this is to

 

 25   reemphasize what Phil said, it is not that we can't

 

                                                               342

 

  1   do it, so that means it shouldn't be done.  It is

 

  2   to try and identify what is critical, so that you

 

  3   make sure that the critical points are done, so

 

  4   that is the critical issue to me.

 

  5             DR. RIEVES:  Dr. Lederman's presentation

 

  6   was excellent, and I think it raised some excellent

 

  7   points.  I think the important part will be to

 

  8   discuss them tomorrow, and can give one example,

 

  9   because for every point that was raised in that

 

 10   discussion, there is always the other hand. It is

 

 11   like the two-armed economist. I will give you one

 

 12   example right now.

 

 13             It is true that one study with what is a

 

 14   laser TMR system, there were very few perforations.

 

 15   There was another blinded, randomized study,

 

 16   completed in the U.S., published in JACC, 140

 

 17   patients.  Only the treated patients, 70 treated

 

 18   patients actually were catheterized.

 

 19             Now, in that study, 5 of them had

 

 20   perforations, so it is often difficult to--just as

 

 21   one example, there is always another side to this,

 

 22   and the important thing is I think you have raised

 

 23   some excellent points.  We have left the tough

 

 24   questions for our committee members to address.

 

 25             DR. SIMONS:  To come back to the point

 

                                                               343

 

  1   that catheters are different, not only are they

 

  2   different from each other, they are different from

 

  3   the cells depending on the cell type used.  I

 

  4   absolutely do not think there could be universal

 

  5   device.

 

  6             DR. LEDERMAN:  And these questions were

 

  7   not answered in benchtop testing?

 

  8             DR. SIMONS:  No.

 

  9             DR. RAO:  I think your point is well made,

 

 10   though, that I think the way devices need to be

 

 11   regulated is somewhat different from cells, because

 

 12   of the number of variables one might have to

 

 13   consider are somewhat different, and I think that

 

 14   is a very valid point.

 

 15             If there is no more questions,  we will go

 

 16   to the open part of the question and answer

 

 17   session.

 

 18                       Open Public Hearing

 

 19             Before we can have the open public

 

 20   hearing, by law, I am required to read a statement.

 

 21   I will do that right now.

 

 22             Both the Food and Drug Administration and

 

 23   the public believe in a transparent process for

 

 24   information gathering and decisionmaking.  To

 

 25   ensure such transparency at the open public hearing

 

                                                               344

 

  1   session of the advisory committee meeting, FDA

 

  2   believes that it is important to understand the

 

  3   context of an individual's presentation.

 

  4             For this reason, FDA encourages you, the

 

  5   open public hearing speaker, at the beginning of

 

  6   your written or oral statement, to advise the

 

  7   committee of any financial relationship that you

 

  8   may have with any company or any group that is

 

  9   likely to be impacted by the topic of this meeting.

 

 10             For example, the financial information may

 

 11   include the company's or a group's payment of your

 

 12   travel, lodging, or other expenses in connection

 

 13   with your attendance at the meeting.  Likewise, FDA

 

 14   encourages you at the beginning of your statement

 

 15   to advise the committee if you do not have any such

 

 16   financial relationship.

 

 17             If you choose not to address this issue of

 

 18   financial relationships at the beginning of your

 

 19   statement, it will not preclude you from speaking.

 

 20             There were two people who had asked to be

 

 21   recognized before the meeting started.  The first

 

 22   person is Dr. Vulliet.

 

 23             DR. VULLIET:  Thank you for the

 

 24   opportunity to come and present some data to you.

 

 25   Am I supposed to make a statement I have no

 

                                                               345

 

  1   financial interest in this?

 

  2             DR. RAO:  Yes.

 

  3             DR. VULLIET:   Okay.  I have no financial

 

  4   interest in this.

 

  5             This is an example of some studies that

 

  6   were done very recently where we have been

 

  7   investigating, using my research team, which is

 

  8   myself, a cell biologist/pharmacologist, Dr.

 

  9   Greeley is a pathologist, Mitch Halloran is a cell

 

 10   biologist, Kristin McDonald and Mark Kittelson are

 

 11   both board-certified cardiologists, so we have a

 

 12   very interdisciplinary team.

 

 13             We are at a vet school, which is probably

 

 14   novel for this group, and we are very specifically

 

 15   interested in animal models.  I was very pleased to

 

 16   hear quite the discussion of animal models.  I

 

 17   disagree with almost everything every one of the

 

 18   speakers complained about, not being suitable

 

 19   animal models.

 

 20             I guarantee we see animal models that

 

 21   definitely are real patients, that have real

 

 22   disease.  It is not induced, it is a real disease.

 

 23   It is there, it needs to be treated.  For that

 

 24   reason, we have decided to investigate the

 

 25   possibility of using cytotherapeutics to see if we

 

                                                               346

 

  1   can produce a beneficial effect in animals.

 

  2             A good example of our animal model--and

 

  3   this slide is probably Peter's example of what they

 

  4   think we do with animals--but this is Oscar, and I

 

  5   guarantee you he will grow up to have somewhere

 

  6   later in life, lumbar disease, lumbar disk disease,

 

  7   either at the L2/3 or the L3/4, and he is a great

 

  8   model if you are into disk disease, but that is a

 

  9   different committee we talk to about that.

 

 10             Steps for successful cytotherapeutics.

 

 11   This is my perception, not the committee's

 

 12   guideline, the first step is safety studies.  This

 

 13   is about where we are at.  In fact, most of the

 

 14   stuff we are squabbling about right now is whether

 

 15   these things can be done safely or not, if you

 

 16   think about it.

 

 17             Very little good data on dose response,

 

 18   nothing on time course that I am aware of.  Nothing

 

 19   or very little on clinical endpoint.  What do I

 

 20   mean by clinical endpoint?  When I am giving an

 

 21   antibiotic, I can tell you I need to hit serum

 

 22   concentrations of 1 microgram per ml.

 

 23             Okay.  I can design a pharmacokinetic

 

 24   regimen, I can hit 1 microgram per ml.  If I am

 

 25   treating dilated cardiomyopathy, what is my

 

                                                               347

 

  1   endpoint?  If I see something four to six weeks

 

  2   later, I will be lucky.

 

  3             So, when I am administering cells, I

 

  4   really don't have a defined clinical endpoint at

 

  5   this point other than the lack of adverse reaction.

 

  6   Think about it.  It is a very interesting point of

 

  7   view.

 

  8             Anyway, because we started off with safety

 

  9   studies, that is what we did.  We asked a very

 

 10   simple hypothesis, and we started with can a half a

 

 11   million cells--we are using mesenchymal stromal

 

 12   cells, we call them stromal cells rather than stem

 

 13   cells because we are not convinced that primordial

 

 14   germ layers have been demonstrated coming from MSC,

 

 15   so we refer to them as stromal cells, you can call

 

 16   them stem cells if you like.

 

 17             We also use a terminology I don't think

 

 18   anybody else has used in this room yet, is we are

 

 19   using MICs [ph]. That is a million cells per

 

 20   kilogram.  We are very interested in a

 

 21   dose-response relationship.  Doses are a key in any

 

 22   therapeutics.

 

 23             So, we are giving half a million MSCs per

 

 24   kilogram of body weight, and can they be safely

 

 25   injected into the coronary arteries of the

 

                                                               348

 

  1   anesthetized dog?

 

  2             Simple experimental design, they are

 

  3   autologous.  We collect bone marrow somewhere about

 

  4   a month later, we inject 10 million cells.  These

 

  5   are 20 kilogram dogs, half-million cells per

 

  6   kilogram.

 

  7             Seven days later, the dogs were

 

  8   anesthetized, physical exam, CV exam, necropsy and

 

  9   histo and immunocytochemistry.

 

 10             I should also point out everywhere in this

 

 11   study, all of these dogs, after recovering from

 

 12   anesthesia, passed the cold nose test.  They were

 

 13   perfectly normal, you would not be able to tell.

 

 14   They jumped, their tails wagged, they licked your

 

 15   hand.  They were nice dogs.

 

 16             So, in that regard, on physical exam, they

 

 17   looked good.

 

 18             Abbreviated methods.  Autologous, four

 

 19   sites, collect them.  Freshly dispersed, and this

 

 20   will be a key point at the end.  These are

 

 21   autologous cells, freshly dispersed MSCs into the

 

 22   circumflex artery.  Catheter placement verified

 

 23   before injection, after injection, with

 

 24   fluoroscopy, physical exam.  All dogs basically

 

 25   appeared to be normal once the effects of

 

                                                               349

 

  1   anesthesia had worn away.

 

  2             This is the first dog we did.  This is the

 

  3   highest dose we did, and we did 1 million cells per

 

  4   kilogram, injected in the coronary.  At 2 hours,

 

  5   took a section, you can see the catheter placement

 

  6   there, took a section of ventricular myocardium.

 

  7   It's a lightly stained hematoxylin section.  These

 

  8   2 cells, basophilic cells here, translate to the

 

  9   CMFDA-labeled cells or fluorescent cells there.

 

 10             This is one of the things we are looking

 

 11   for.  Our research team, probably different than

 

 12   many of the people in this room, feel that the only

 

 13   way we will get effective therapeutics is to have

 

 14   intimate contact between these cells and the dead

 

 15   and dying cardiomyocytes that they are going to

 

 16   replace.  We don't believe in direct injection, and

 

 17   we can talk about that later.

 

 18             So, that was our first dog, 1 million

 

 19   cells per kilogram.  The reason he went--it was at

 

 20   two hours--was he went into V fib and died.  Two

 

 21   hours, so it is fairly easy to do your necropsy in

 

 22   that.  So, we then bacted those off  to a

 

 23   half-million cells per kilogram and injected them,

 

 24   and the injections are 5 injections of 2 ml each

 

 25   and 1 million cells per ml, and this was the

 

                                                               350

 

  1   control, the anesthetized dog, this is the ECG.

 

  2             Dogs normally have inverted T waves, I

 

  3   don't know how many people know that, and it has to

 

  4   do with chest dimensions and chest geometry more

 

  5   than anything else.  This is a normal ECG for a

 

  6   dog.

 

  7             After the fifth injection, the T wave has

 

  8   converted to a normal position, but more

 

  9   importantly, what you see here is ST elevation,

 

 10   very profound ST elevation. This increase with each

 

 11   dose and at the fifth injection it was the most

 

 12   severe.

 

 13             Twenty-four later, post-injection, you can

 

 14   see you have got back to an inverted T wave, but

 

 15   you have got bizarre complexes here.  One-week

 

 16   post-injection you have got normal ECG, and this

 

 17   was published recently in Lancet, so I am not going

 

 18   to go through a lot of this published thing two

 

 19   weeks ago.

 

 20             ST elevation made us think of troponin.

 

 21   We measured troponin at various times after

 

 22   injection.  You can see it goes up, increases to

 

 23   about 45 nanograms per ml.  If you ligate an LAD in

 

 24   a dog, you normally see ranges in the order of 150

 

 25   to 200, so we had subnormal, if you will, levels of

 

                                                               351

 

  1   troponin, but the time course it would be

 

  2   consistent with some sort of myocardial ischemia.

 

  3             One of the things you also don't see in

 

  4   many of these preparations that you are seeing,

 

  5   which is my personal--I will give you guys my

 

  6   personal things that sort of irk me a little bit

 

  7   about science--is we are using H&E histology.

 

  8             This is the gold standard of pathology.  I

 

  9   don't care how many immunocytochemistry studies you

 

 10   see, H&E is what medical pathologists use to

 

 11   evaluate an outcome of a case.  It is essential, it

 

 12   should be included, I don't know why people don't

 

 13   use it, but it is a great technique, I like it

 

 14   although I didn't do very well in pathology.

 

 15             Here is a good example of a section of the

 

 16   ventricle of one of the injected dogs.  What you

 

 17   see is three areas here of hypercellularity.  This

 

 18   is the normal myocardium here as you zoom in.  This

 

 19   is at 4X or 40X, this is 100X and 400X, and we are

 

 20   zooming in on this area right in here.

 

 21             You see it is more hypercellular.  What

 

 22   you see in here is you see mononuclear cells,

 

 23   rounded nucleus.  You see some elongated nucleus.

 

 24   You see fibrosis.  You see some lytic lesions.  I

 

 25   looked at that as a cell biologist, and I said,

 

                                                               352

 

  1   great, that's where my stem cells are, right?

 

  2   Wrong. My pathologist looked at it and said, "Rick,

 

  3   you have got a problem.  Those are macrophages.

 

  4   You have just produced a heart attack in this dog."

 

  5   And I said, "Oh."

 

  6             So, to verify that, these were CM dye/I

 

  7   labeled cells zooming in.  You can see that the CM

 

  8   dye/I label is in the vicinity of this

 

  9   hypercellularity, so both the MSCs and these

 

 10   macrophages are here, but how do we know they are

 

 11   macrophages?

 

 12             Again, using canine-specific antibodies

 

 13   that we have available in the vet teaching

 

 14   hospital, this is an H&E section, this is a CD18

 

 15   monoclonal antibody specifically raised against

 

 16   canine macrophages.  You can see you have punctate

 

 17   lesions, it is very characteristic, and, indeed, to

 

 18   confirm this, 7 days later, what we also see

 

 19   characteristic of myocardial infarctions is

 

 20   increased fibrosis and collagen deposition.

 

 21             As you can see here, this is normal

 

 22   myocardium, it stains very red in Masson's, and

 

 23   what you see here is you see the blue here is

 

 24   collagen fiber deposition.  This is very

 

 25   characteristic of myocardial infarction.

 

                                                               353

 

  1             Five cardinal signs of MI are ECG changes,

 

  2   we showed that; proteins released from damaged

 

  3   myocardium, we showed that; decreased wall motion,

 

  4   we did not see that.  We did ultrasound, but I

 

  5   don't know that we could.  We have the

 

  6   sophistication to measure wall motion.  Myocardial

 

  7   infarction is just not a common veterinary disease

 

  8   that we see.  Characteristic cellular infiltrates,

 

  9   we saw that.  Collagen deposition.

 

 10             Basically, our conclusion is that at 0.5

 

 11   million cells per MSC, will produce myocardial

 

 12   ischemia, microinfarctions in these dogs.

 

 13             Our original interpretation of this was

 

 14   that this was a dose/rate of delivery problem.  So,

 

 15   our feeling was, because there really is not much,

 

 16   is you critically look at the clinical studies out

 

 17   there in humans, they have got doses all over the

 

 18   place.  It is very hard to extrapolate what the

 

 19   dose is.  That is why use milligrams per kilogram.

 

 20   I would encourage anybody doing these studies to

 

 21   use some sort of a normalization like that.

 

 22             I would encourage the committee to require

 

 23   it, so that you can start comparing, but more

 

 24   importantly, what we did, just to give you an in

 

 25   idea, is possibly post-injection cell clumping.  We

 

                                                               354

 

  1   didn't consider this as a possibility.  We have

 

  2   taken the holding media the cells were in, and the

 

  3   injection media, to inject them into the cells.

 

  4             At the end of 2 hours of holding them in

 

  5   the holding media, I personally inspected most of

 

  6   the dogs.  We inspected the cells.  None of the

 

  7   cells are clumped, so we assumed clumping was not a

 

  8   problem.

 

  9             Just to give you guys a little bit more

 

 10   gray hair in terms of your job as far as making

 

 11   decisions, what we didn't do is we didn't do the

 

 12   right control, and the right control was to put

 

 13   these cells in 100 percent serum, because that is

 

 14   what you are doing, you are injecting them into the

 

 15   artery.  From the time they leave that artery, they

 

 16   are going into 100 percent serum.

 

 17             We did that and the cells started

 

 18   clumping.  I don't know why.  They didn't clump

 

 19   when we pulled them out of the bone marrow because

 

 20   they would have been clumped when we put them in

 

 21   tissue culture dish.

 

 22             They didn't clump then, but during the

 

 23   process to  2 weeks of preparing them, sticking to

 

 24   plastic, their adhesive properties had changed.

 

 25   One dog, and I have heard several people talk about

 

                                                               355

 

  1   fetal bovine serum, I don't know if it is known to

 

  2   this group or not, Darlen Procoff [ph] last year

 

  3   had an abstract, I don't think he has published it

 

  4   yet, had an abstract where he looked at fetal

 

  5   bovine serum in cells.  It carries over, I believe

 

  6   it was about a milligram per million cells if you

 

  7   grow cells in FBS, at least with the MSCs, will

 

  8   pinocytose and hold about a milligram per million

 

  9   cells of fetal bovine serum.

 

 10             It is not released by washing.  These

 

 11   cells were rinsed three times in Hanks' balanced

 

 12   salt before they were injected.  If you want to

 

 13   remove the fetal bovine serum, you have to grow

 

 14   them for at least 48 hours to get rid of it.

 

 15             This may explain some of the early dieoff

 

 16   of the myoblasts that you are seeing, if you have

 

 17   got FBS in there, because you will get a reaction

 

 18   from it.

 

 19             Basically, this research team does not

 

 20   feel that we are in a position to perform these

 

 21   studies on client-owned patients at this time,

 

 22   although that is our long-term goal.

 

 23             What do we think is happening?   What we

 

 24   think is happening is, we think the cells are

 

 25   coming in, we think they are clogging, producing

 

                                                               356

 

  1   microinfarctions by clogging either a second or

 

  2   third order arterial, and causing areas of ischemia

 

  3   and microinfarctions.

 

  4             Are there clinical techniques to detect

 

  5   this?  That is one of those things.  Would you see

 

  6   that, or does all the old imaging, does the modern

 

  7   world ultrasound pick up something like that?  I

 

  8   don't think so.  So, that is why I am saying

 

  9   histology is pretty important.

 

 10             How do our cell preparations compare?

 

 11             DR. RAO:  Dr.  Vulliet, we would like try

 

 12   and make sure that we stay on time.

 

 13             DR. VULLIET:  Sure.  Let me finish this

 

 14   because that is actually what I think we are more

 

 15   interested in.

 

 16             This is bone marrow from canine bone

 

 17   marrow.  As you can see, you have got metas and

 

 18   milas and bands, and that kind of stuff.  This is

 

 19   canine MSCs.  Our cell size is about 19 microns, 20

 

 20   microns.  Mean cell size on this population,

 

 21   because they are much smaller, is 10 to 12 microns.

 

 22             More importantly, look at this.  The range

 

 23   on these, again, these are characteristic things.

 

 24   These are done in dog, they don't publish them in

 

 25   humans.  Range is 7 to 50 microns, okay, and it

 

                                                               357

 

  1   could go even go higher, go as high as 80.

 

  2             Plasticity in this population of cells,

 

  3   this is probably being generous, saying it is about

 

  4   0.01 percent.  If you take the canine population of

 

  5   cells, and you go through a CFU selection process,

 

  6   you can get plasticity on the order of 40 to 60

 

  7   percent.

 

  8             If you are starting to compare potency,

 

  9   potential  potency between these two preparations

 

 10   of cells, this preparation of cells, after it went

 

 11   through a CFU selection process, would be about

 

 12   4,000 to 40,000 times more potent than just crude

 

 13   bone marrow in terms of plasticity potential, if

 

 14   you will.

 

 15             Intrinsic properties of these cells, I

 

 16   don't know. These cells have a tendency to lay down

 

 17   collagen if you just leave them sitting in a tissue

 

 18   culture dish.

 

 19             Successes for safety studies,

 

 20   therapeutics, efficacy studies need to be done in

 

 21   well characterized model diseases and patient

 

 22   diseases.

 

 23             What I would like to do is leave you with

 

 24   a couple of philosophies.  We believe that bone

 

 25   marrow stem cells have potential to treat many cell

 

                                                               358

 

  1   loss diseases, especially the myocardium, and we

 

  2   will continue using these even in spite of the

 

  3   negative report.  We believe this is a technical

 

  4   problem.  We believe we will solve it by adjusting

 

  5   the dose and a few other things.

 

  6             However, strict scientific disciplines are

 

  7   necessary to avoid a train wreck.  As Doris Taylor

 

  8   said, we do not want to repeat the gene therapy

 

  9   trial.

 

 10             The other thing I can give you is those of

 

 11   us who used to have gray in our hair, and now I

 

 12   don't have hair, is the comment, when clinicians

 

 13   read their own press clippings, patients are going

 

 14   to suffer.

 

 15             Thank you for your attention.

 

 16             [Applause.]

 

 17             DR. RAO:  Thank you, Dr. Vulliet.

 

 18              Our next speaker is from Genzyme.  Let's

 

 19   try and restrict the time to about 10 minutes.

 

 20             MR. DU MOULIN:  It is very tough being the

 

 21   last speaker of the day, but it is absolutely worse

 

 22   being the last speaker of the day and following a

 

 23   vet who talks about puppies.

 

 24             [Slide.]

 

 25             Good afternoon.  My name is Gary C. du

 

                                                               359

 

  1   Moulin.  I am vice president of Quality Systems for

 

  2   the cell therapy operations at Genzyme Corporation

 

  3   in Cambridge, Massachusetts.

 

  4             Genzyme Corporation is collaborating with

 

  5   Professor Philippe Menasch of Paris, France, in a

 

  6   multicenter, Phase II/III clinical study autologous

 

  7   skeletal myoblast implantation in Europe.

 

  8             [Slide.]

 

  9             Ensuring the therapeutic success of

 

 10   cardiac cell therapy is predicated on a rigorous

 

 11   scalable autologous cell culture program based upon

 

 12   the principles and practices of good manufacturing.

 

 13             Our long experience with the scale-up and

 

 14   delivery of cartilage and keratinocyte-based cell

 

 15   therapy products and services to thousands of

 

 16   patients has confirmed that each element of good

 

 17   manufacturing practices contributes an essential

 

 18   part of an overall program that optimizes chances

 

 19   of providing cell therapy products expressing the

 

 20   attributes of safety and consistent quality for

 

 21   patients.

 

 22             Controls required for the manufacturing

 

 23   process begin at the collection site of the muscle

 

 24   biopsy and ends approximately three weeks later as

 

 25   the suspension of cells exhibiting quality and

 

                                                               360

 

  1   safety characteristics that once implanted can

 

  2   consistent initiate a robust repair process.

 

  3              Maintaining the sterility of the cell

 

  4   culture system and ensuring lot segregation are

 

  5   critical attributes of success.

 

  6             [Slide.]

 

  7             All the elements of the GMP-based

 

  8   manufacturing program are essential in order to

 

  9   control the inherent variability representative of

 

 10   autologous cell culture. Briefly, these controls

 

 11   must be established based upon the following

 

 12   aspects of GMP.

 

 13             These include, and they are listed here, a

 

 14   process that is validated, personnel who are

 

 15   trained and certified to manipulate cell safely, an

 

 16   appropriate facility expressing stringent

 

 17   environmental controls, records and documentation

 

 18   of all processes conducted, equipment that is

 

 19   calibrated and validated, raw materials that have

 

 20   been tested for their quality, accepted formally,

 

 21   and released into the manufacturing stream.  Unique

 

 22   to an autologous cell process is to maintain

 

 23   stringent patient lot segregation.

 

 24             [Slide.]

 

 25             This is a photograph of our sole

 

                                                               361

 

  1   manufacturing facility.  It is approximately 10,000

 

  2   square feet, contains about 70 biosafety cabinets

 

  3   in which individual patient's tissues are

 

  4   manipulated, but other organizational requirements

 

  5   necessary beyond the GMPs include these elements

 

  6   here, beyond the manufacturing, a purchasing

 

  7   element, materials handling element, logistics for

 

  8   the shipment of cells, customer care for

 

  9   communicating with the surgeon and the patient,

 

 10   engineering and facilities to maintain your

 

 11   facility, manufacturing technical services

 

 12   responsible for the training of personnel, process

 

 13   development, and clinical manufacturing, a

 

 14   formalized quality assurance, quality control, and

 

 15   validation services program.

 

 16             [Slide.]

 

 17             In order to scale up manufacturing

 

 18   activities for a clinical development paradigm, an

 

 19   organization must be created to effectively manage

 

 20   a myriad of direct and ancillary responsibilities,

 

 21   but here, quality controls including environmental

 

 22   monitoring for the manufacturing facility are

 

 23   critical components of the operational elements of

 

 24   cell therapy productions.

 

 25             Robust testing programs ensures--and I

 

                                                               362

 

  1   have listed them here--that cell therapy products

 

  2   meet the highest standards of safety,

 

  3   effectiveness, and reliability as a therapeutic

 

  4   modality, that transmission of communicable

 

  5   diseases is prevented, that one ensure that all

 

  6   manufacturing and processing controls are in place

 

  7   and consistently followed, that there is compliance

 

  8   with existing and anticipated regulatory

 

  9   requirements, that validated assays are performed

 

 10   which support lot release and performance

 

 11   monitoring of materials and components, and,

 

 12   finally, encouraging the development of new assays

 

 13   which enhance product safety, and, finally,

 

 14   generating and analyzing that data, the

 

 15   quantitative data to support continuous

 

 16   improvements to your process.

 

 17             [Slide.]

 

 18             Putting these concepts together, a

 

 19   manufacturing process whose key manufacturing

 

 20   events from biopsy receipt through final product

 

 21   fill finish are well understood, can optimize the

 

 22   ex vivo cell culture process, and supported by

 

 23   validated quality controls can ensure safety and

 

 24   product consistency.

 

 25             [Slide.]

 

                                                               363

 

  1             We believe that call product

 

  2   characterization is possible based upon validatable

 

  3   measures of viability, purity, identity, and yield,

 

  4   with safety indicators of sterility, endotoxin in

 

  5   the absence of mycoplasma.

 

  6             Here are shown the percent viability and

 

  7   percent CD56 expression of three cell therapy

 

  8   products prepared during process validation studies

 

  9   from cadaveric skeletal muscle tissues.

 

 10             Included in these data is evidence of cell

 

 11   product stability over a 72-hour time frame, one

 

 12   reasonably to be expected if transportation of the

 

 13   cells over a long distance to the patient is

 

 14   required.  Note that the lot release parameters

 

 15   remain stable over 72 hours.

 

 16             Viability and CD56 expression, both flow

 

 17   cytometric and validatable measures of cell product

 

 18   identity can be consistently maintained above the

 

 19   90 percent range.

 

 20             [Slide.]

 

 21             Measures of sterility and potency are

 

 22   shown in this slide.  It is crucial to ensure that

 

 23   sterility is maintained throughout the

 

 24   manufacturing process.  In the case of short shelf

 

 25   life cell therapy products utilizing automated

 

                                                               364

 

  1   microbial detection systems can provide benefits to

 

  2   improve time to detection should microbial

 

  3   contaminants be present.

 

  4             Finally, in order to demonstrate the

 

  5   potential for a therapeutic effect, the presence of

 

  6   myotubule formation can be used as a measure of

 

  7   identification and perhaps potency.  Myoblasts are

 

  8   undifferentiated muscle precursor cells which, when

 

  9   fused, become the differentiated myotubules.  The

 

 10   presence of multinucleated muscle cells is a strong

 

 11   visual indicator that muscle differentiation has

 

 12   occurred and may be an important predictor of cell

 

 13   function.

 

 14             In these photographs, one can readily see

 

 15   multinucleated myotubules indicated by the yellow

 

 16   arrows from freshly prepared samples and in cells

 

 17   after a simulated 72-hour shipment period.

 

 18             [Slide.]

 

 19             In conclusion, quality, safety, and

 

 20   effectiveness must be designed into cell therapy

 

 21   products.  Quality cannot be inspected in or tested

 

 22   into cell therapy products.  Despite the fact that

 

 23   we may, at this time, not know every aspect of cell

 

 24   product characterization, institution and

 

 25   maintenance of stringent manufacturing controls

 

                                                               365

 

  1   through rigorous observance of GMPs can contribute

 

  2   to the safety and consistency necessary for the

 

  3   production of cell therapy products intended for

 

  4   cardiac cell therapy.

 

  5             Developers of cell therapy products must

 

  6   consider, understand, and incorporate quality

 

  7   requirements at the earliest possible stages of the

 

  8   clinical development program, and in so doing, can

 

  9   optimize the therapeutic potential of these

 

 10   promising technologies.

 

 11             Thank you.

 

 12             [Applause.]

 

 13             DR. RAO:  Thank you.

 

 14             If there are no questions, I would like to

 

 15   ask if anybody else from the audience wishes to

 

 16   make any comment at this time.  I am going to ask

 

 17   that they limit their comments and be brief.

 

 18             [No response.]

 

 19             DR. RAO:  If there are no more comments,

 

 20   then, I can declare the meeting adjourned.

 

 21             [Whereupon, at 5:46 p.m., the proceedings

 

 22   were recessed, to reconvene at 8:00 a.m., Friday,

 

 23   March 19, 2004.]

 

 24                              - - -