Title of Invention

"A METHOD FOR DIRECTING THE DIFFERENTIATION OF PRIMATE EMBRYONIC STEM CELLS INTO ENDOTHELIAL CELLS"

Abstract A method is described to induce primate embryonic stem cells to differentiate into a relatively homogenous population of endothelial cells. The ES derived endothelial cells have the general morphological and cell surface marker characteristics of endothelial cells. The ES derived endothelial cells also are capable of inducing and participating in blood vessel formation (or vascularization) when transplanted into tissue in vivo. (FIG. NIL)
Full Text A METHOD FOR DIRECTING THE DIFFERENTIATION OF PRIMATE
EMBRYONIC STEM CELLS INTO ENDOTHELIAL CELLS
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority from U. S. Provisional Patent Application No.
60/335,332 filed November 2,2001.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
[0002] To be determined.
BACKGROUND OF THE INVENTION
[0003] Stem cells are defined to be cells which are capable both of self-renewal and
differentiation into one or more differentiated cell types. Human embryonic stem cells are a
category of stem cells created from human pre-implantation blastocysts. Human embryonic stem
cells are pluripotent and may be totipotent, meaning that they can certainly differentiate into
many cell types evidenced in an adult human body and may be capable of differentiating into all
cell types present in the human body.
[0004] Embryonic stem cells (ES cells) have also been derived in a number of animals
other than humans. For example, much scientific work has been conducted with murine ES cells.
Once a method for the initiation of ES cell cultures for a particular species is worked out, it
becomes possible to manipulate the ES cells, and animals which result therefrom, in a variety of
ways to learn useful information about the genetics of the animal under study. For example, it
has become possible over the past decade to create cultures of murine ES cells in which one or
another specific gene is knocked out in each murine stem cell culture. While some techniques
that could be worked out in murine ES cell systems were transferable to other species, many
were not. For example, the basic techniques which could be used to create murine ES cell
cultures did not transfer well to many other animal species. For the development of techniques
for the culture and manipulation of human ES cells, the murine cell may therefore not be the best
model due to the phylogenic distance between humans and mice. However, in the course of the
development of the science of human ES cell cultures and techniques, much of the preliminary
work was conducted in non-human primates, such as the rhesus monkey. Other primate ES cell
cultures have proven to be a relatively reliable model for systems that could be easily transferred
to human cell culture. For an example, murine ES cell cultures require application of leukemia
inhibitory factor (LIF) or another agonist of the gpl30/STAT3 signaling pathway for
maintenance of undifferentiated cell growth, whereas human and rhesus monkey ES cell cultures
do not require LIF for undifferentiated cell growth. Prior work on hematopoiesis using rhesus
monkey ES cells validates the utility of this system for doing pre-clinical investigations for
techniques mat can be transferred to human ES cell cultures.
[0005] One of me exciting potential uses of stem cells is for human tissue transplantation.
It is hoped and expected that techniques can be developed to direct the differentiation of stem
cells into specific lineages which can then be transferred into the human body to replace or
enhance tissues of the body. In order to do that, first techniques must be developed to direct the
differentiation of stem cells into the specific cell lineages desired. Techniques have already been
proposed to direct stem cell cultures into lineages of hematopoeic, neural, cardiomyocyte,
pancreatic and other lineages. These techniques have proven to be quite different from each
other and independent in the sense that a new and different technique is required for each new
desired lineage.
[00061 Endothelial cells make up a network of interconnected cells in the human body
that line blood vessels, lymphatic vessels, and form capillaries. Endothelial cells regulate the
flow of nutrient substances and create and respond to diverse biologically active molecules.
While it has been demonstrate that human ES cells will differentiate into many progeny cells
types, including endothelial cells, it has not been previously possible to create distinct cultures of
derivatives of human ES cells directed into an endothelial lineage.
BRIEF SUMMARY OF THE INVENTION
[00071 The present invention is summarized in that a method has been developed which
permits the direct differentiation of a culture of embryonic stem cells into a culture of endothelial
cells. The method includes culturing the embryonic stem cells in a culture medium previously
known to maintain endothelial cells and which, it now turns out, has the capability to support
embryonic stem cells in the process of differentiation into endothelial cells.
[0008] The present invention is also summarized in that cultures of endothelial cells
derived from embryonic stem cells which have morphology and cell surface markers
characteristic of endothelial cells and which are capable of inducing vascularization of tissue

vivo-
[0009] it is a feature of the present invention in that it is relatively efficient to perform
since its steps are simple, and the result is a culture that appears to be a relatively homogenous
population of endothelial cells.
[00010] Other objects, advantages and features of the present invention will become
apparent from the following specification when taken in conjunction with the accompanying
drawings.
BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWINGS
Not applicable.
DETAILED DESCRIPTION OF THE INVENTION
[00011] The Present invention is directed both to a method to direct the differentiation of
primate embryonic stem cells into endothelial cells and to the relatively pure population of
endothelial cells so produced. The method is based on the cultivation of primate embryonic stem
cells with a defined protein growth factor or factors which cause the cells so treated to change
their morphology to become endothelial cells. In contrast to other techniques for the directed
differentiation of cells of other lineages from embryonic stem cells, the culture of endothelial
cells, derived from embryonic stem cells by the method described here, appears relatively
uniform and is made up of primarily of endothelial cells having apparent angiogenic capability.
[00012] The culture method is based on the culture of undifferentiated primate embryonic
stem (ES) cells in a medium containing vascular endothelial cell growth factors (VEGF), basic
fibroblast growth factor (bFGF), insulin-like growth factor (IGF-1), and epidermal growth factor
(EGF). These factors are all found in a commercially available medium known as endothelial
cell basal medium (EBM-2, Clonetics/BioWhittaker). This medium was previously known and is
used to sustain endothelial cells in culture. It was not previously known that this medium could
be used to support the differentiation of ES cells into endothelial cells. While this combination
of growth factors has been found to be sufficient to support the differentiation of ES cells into
endothelial cells, it may not be necessary to use all four factors in the culture medium, and
whether or not one of the factors can be omitted can readily be ascertained by empirical
experimentation without departing from the concept of the present invention.
[00013] What separates this method from prior art derivation of heterogeneous mixtures
including endothelial cells is the relative uniformity of the transition of the cell culture from ES
cells to endothelial cells. Other methods were tried, without success, to achieve this transition,
such as application of phorbol esters, co-cultivation with stromal cells plus serum and isolation of
endothelial cells from embryoid bodies. None of these efforts reproducibly yielded cultures of
predominantly endothelial cells. In contrast, the method described here is simple and efficient
and results in a cell culture of morphologically similar cells having the characteristics of
endothelial cells.
[00014] The culture of endothelial cells made by the present invention will have certain
characteristics. The cells have a characteristic morphology, similar to elongated or stellate
shaped endothelial cells. In contrast, ES cells grown in other media differentiate into a
heterogeneous population of cell types with no distinct endothelial-appearing cells. The
endothelial cells rapidly form tubular structures when placed in Matrigel (™) medium. The
endothelial cells are positive for presence of the von Willebrand factor (vWF) and have high
levels of ulex europaeus agglutinin 1 (UEA-1) binding, as well as expression of the intergrin
avß3 and the surface antigen CD 146. These cells also will take up acetylated LDL, another trait
characteristic of endothelial cells. These cells do lack expression of CD31 and VE-cadherin, two
antigens commonly, but not always, present on the surface of endothelial cells. These
endothelial cells have the ability, when transferred into a SCID (severe combined
immunodeficient) mouse together with tumor cells, to effect the vascularization of the resulting
tumor, thus demonstrating the ability of the cells both to recruit and to participate in
vascularization in vivo. The ability of these cell to participate in vascularization is particularly
noteworthy, since that attribute makes in possible to transplant genetically altered endothelial
cells into a tissue requiring vascularization with the altered cells surviving in vivo in the vascular
matrix created to therefore express whatever gene was inserted into the cells.
[00015] In contrast to other cell types which can be induced to form from embryonic stem
cells, the endothelial cell culture described and characterized here is relatively homogenous in
cells committed to the derivative lineage, i.e. to be endothelial cells. The ES derived endothelial
cell culture is formed of cells having a uniform morphology and exhibiting the characteristics of
endothelial cells. Given the limits of present cell culture technology, however, it cannot be said
with certainty that the ES derived endothelial cell culture is entirely free of other cell types.
What can be said is that the ES derived endothelial cell culture is predominantly composed of
endothelial cells and is a practical source of cells which will act as endothelial cells to promote
and participate in vascularization of tissues when transplanted into a host in vivo- Using a
common test for endothelial character, the ability to bind the Ulex europaeus agglutinin 1 (UEA-
1) lectin, it has been found that reproducible over 90% of the cells in the derivative culture do
bind the UEA-1 lectin. While in some variations of the method the percentage of cells which
bind to UEA-1 might vary, in cultures of endothelial cells made by the method described here, at
least 75%, and more preferably, over 90%, of the cells in the culture will test positive for the
ability to bind the UEA-1 lectin.
[00016] While the examples below were conducted with rhesus monkey ES cells, the same
processes and result can be obtained with human ES cells. Human endothelial cells derived from
ES cells offer the possibility to develop tissues transplantable into human patients.
Transplantation of endothelial cells would be desirable for those applications in which
vascularizarion of ischemic tissue is needed. In addition, the introduction of endothelial cells
may be useful in any location on the body where improved vascularization is needed. Since the
precursor ES cells can be grown in any number, this makes possible the generation of large
numbers of endothelial cells for clinical experimentation or treatment.
EXAMPLES
[00017] METHODS
[00018] Cell Culture
[00019] Undifferentiated rhesus monkey ES cells (R366.4 cell line) were cultured as
previously described (Thomson et al. Proc.Natl. Acad. Sci. USA 92:7844-7848 (1996)). Briefly
R366.4 cells were co-cultured with irradiated mouse embryonic fibroblast (MEF) cells in
medium containing DMEM, 20% FBS (Hyclone, Ogden UT), 2mM L-glutamine (Sigma, St.
Louis, MO), O.1mM 2-mercaptoethanol (Sigma), and 1% MEM non-essential amino acids
(Invitrogen). Undifferentiated cells were fed daily with fresh medium and passaged onto new
MEFs approximately every 5-7 days. To promote endothelial cell differentiation, the medium
was removed from the ES cells 24 hours after plating and replaced with medium consisting of
EGM2, 5% FBS, VEGF, bFGF, IGF-1, EGF, and ascorbic acid (EGM2) (EGM2-MV Bullet Kit,
Clonetics/BioWhittaker, Walkersville, MD). The ES cells were differentiated for 29 days in the
EGM2 medium, which was changed every 3-5 days. Differentiated rhesus ES cells, were
dissociated with 0.05% trypsin/0.53mM EDTA (GIBCO/BRL) for 5 minutes, centrifuged, and
re-plated in EGM2 in 10 cm tissue culture dishes without irradiated MEF cells. After 24 hours
non-adherent cells were removed and adherent cells were fed fresh medium. The rhesus ES cell-
derived endothelial cells (RESDECs) could be grown to confluence and serially passaged and
expanded in the EBM2 medium.
[00020] Human umbilical vein cells (HUVECs) (Clonetics/Biowhittaker) were also grown
and passaged in EGM2 by known methods.
[00021] Tube Formation on Matrigel
[00022] 0.2ml of Matrigel (Becton Dickinson) was added to each well of a 24 well tissue
culture plate and allowed to solidify at 37°C for at least 30 minutes. Following gelation, 0.2ml of
a cell suspension containing 5 x 104 -1 x 105 RESDECs was placed on top of the Matrigel. The
cultures were incubated at 37°C/5% CO2 and observed at 24,48, and 72 hours for rearrangement
of cells into tube-like capillary structures. Individual experiments were performed in triplicate
and representative wells recorded by photomicrography.
{00023] VEGF and bFGF ELISA
[00024] RESDECs were cultured for 3 days in the absence of VEGF or bFGF in EGM2,
EGM2 supplemented with 10% Knockout serum replacer (GIBCO) instead of FBS, or DMEM
supplemented with 10% FBS. After 72 hours the conditioned media (CM) was collected and
centrifuged to remove dead cells. EGM2 medium alone served as a negative control. The
amount of VEGF or bFGF in the CM was analyzed by colorimetric ELISA assay (R & D
systems, Minneapolis, MN).
[00025] Flow Cytometry
[00026] RESDECs were washed with Ca2+ and Mg2+ free PBS and detached from the
monolayer with 0.05% trypsin/0.53mM EDTA for 5 minutes. The dissociated cells were
centrifuged and washed with FACS medium consisting of PBS supplemented with 2% FBS and
0.1% sodium azide. After filtration through 80-micron nitex, the single-cell suspension was
measured in aliquots and stained with either isotype control or antigen-specific antibodies diluted
to appropriate concentrations in FACs media. Cell surface antigen expression was analyzed
using antigen-specific primary antibody followed by fluorescent-tagged secondary antibodies
(indirect staining), or fluorescently-conjugated antigen-specific antibodies (Direct staining).
Appropriate unconjugated mouse and goat IgGs (both Sigma) as well as FITC-conjugated mouse
IgG (Pharmingen San Diego, CA)) were used as isotype controls. Unconjugated antigen-specific
antibody against flk-1 (Research Diagnostics) was detected with a FITC labeled anti-goat IgG
antibody (Sigma). Unconjugated antibodies against VEGF receptor 1 (Flt-1) and VEGF receptor
2 (flk-1) (both Sigma) were detected with a FITC labeled goat anti-mouse IgG (Caltag).
Unconjugated P1H12 antibody (mouse IgGl, provided by Dr. Robert Hebbel, University of
Minnesota) was detected with rat anti-mouse IgG-FITC conjugated secondary antibody (Caltag).
The cells were also tested for expression of the VEGF receptor using a biotinylated VEGF Kit
(R&D Systems) and for their ability to bind the ulex europaeus agglutinin 1 (UEA-1) (Vector
labs). Direct conjugated antibodies used were HLA-A, B, C-FITC (Phanningen) and aVp3/cl
LM609-FITC (Chemicon). Human umbilical vein cells (HUVEC) (Clonetics) served as a
positive control. Cells were analyzed without fixation on a FACScan or FACs Calibur (Becton
Dickinson) using propidium iodide to exclude dead cells. Data analysis was carried out using
CellQuest software (Becton Dickinson).
[00027] Immunostaining
[00028] Analysis for the acetylated LDL receptor was performed by diluting dilAcLDL
(Molecular Probes) in serum-free EGM2. Cells were washed twice and incubated overnight in
EGM2 medium containing dilAcLDL. After washing, the cells were observed by fluorescence
microscopy (UV, rhodamine filter). HUVECs were used as a positive control.
[00029] Expression of vonWillebrand factor protein (vWF) (DAKO) was detected with a
goat anti-rabbit IgG-FITC secondary antibody (Sigma). Cells were fixed and incubated at room
temperature for one hour with vWF, washed, and incubated for 30 minutes in the secondary
antibody. After a final wash, cells were observed by fluorescence microscopy (UV, FITC filter).
HUVECS again served as a positive control.
[00030] Matrigel Plugs
[00031] In one experiment SCID mice (Balb/Scid, Harlan Sprague Dawley) were injected
subcutaneously with 0.5ml Matrigel (Becton Dickinson) containing 5 x 105 -1 x 106 RESDECs.
A second experiment was performed implanting a sponge containing the RESDECs into the
solidified Matrigel. The Matrigel plugs were removed after 14,21,35, and 42 days. Vessels
were observed by injecting high molecular weight FITC-dextran (Sigma) intravenously a few
minutes before removing and fixing the plugs. Standard H/E slides were also prepared.
[00032] In Vivo Studies
[00033] Adherent RESDECs were harvested by trypsinization and mixed with the mouse
mammary carcinoma, C755 cell line. In two separate experiments Balb/c-SCID mice were
injected subcutaneously with either 1 x 106 C755 tumor cells, or 1 x 106 RESDECs, or with a
mixture of 1 x 106 tumor cells and 1 x 106 RESDECs. After initial growth, tumors were
measured by caliper every 3-5 days. Approximately three weeks after transplantation all mice
were sacrificed for histochemical analysis of the grown tumors. Mice injected with only
RESDECs failed to grow tumors.
[00034] Immunohistochemical Staining of Tumors
[00035] In the first experiment tumors were isolated and fixed in 10% formalin for the
preparation of paraffin sections. To prepare frozen sections tumors were fixed in 2%
paraformaldehyde. All sections were mounted onto Fisher Superfrost slides. Using the standard
ABC technique (VectaStain Elite ABC kit, Vector labs) sections were processed for expression
of HLA -class IA 3, C (W6/32 antibody, IgG2a) and CD31 (IgG1) (Novacastra, Vector). Mouse
IgGl (Sigma) and IgG2a (Southern Biotechnology) were used as isotype controls. The
peroxidase activity was visualized with a DAB substrate (Vector) and sections were
counterstained with hematoxylin.
[00036] RESULTS
[00037] Derivation of endothelial cells from rhesus monkey ES cells:
[00038] Rhesus monkey ES cells were grown in EGM2 medium containing VEGF, bFGF,
EGF, and IGF as described in materials and methods. After approximately 5-10 days, these ES
cells assumed a uniform morphology similar to elongated or stellate -shaped endothelial cells.
In contrast, ES cells grown in medium supplemented with FBS alone differentiated into a
heterogeneous cell population with no distinct endothelial-appearing cells. The potential
endothelial cells were serially passaged and expanded for approximately 20 population doublings
while grown in EGM2 with maintenance of a homogeneous appearance. As an initial test of
endothelial cell characteristics, these cells were placed in Matrigel-based medium where they
rapidly formed tube-like capillary structures similar those formed by HUVEC or other
endothelial cell populations when placed in Matrigel-based medium. Cytogenetic studies showed
all cells have a normal rhesus monkey 40 XY karyotype. These cytogenetic results were
important to demonstrate that these cells were not transformed after prolonged culture, nor were
they derived from potentially contaminating mouse embryonic fibroblast cells that are used for
the growth of undifferentiated ES cells.
[00039] Electron micrographs of the rhesus ES cell-derived cells demonstrate typical
endothelial cell features. These include multiple dense round or rod-shaped Weibel-Palade
bodies, tight junctions between cells, and endocytic/exocytic vesicles.
[00040] Immunophenotyping
[00041] Next, immunohistochemical staining of these rhesus-derived endothelial-like cells
demonstrated the presence von Willebrand factor (vWF), the ability for these cells to rapidly
take-up acetylated LDL and the ability to bind the UEA-1 lectin. Flow cytometric studies
confirmed high levels of UEA-1 binding, as well as expression of the integrin avj33 and the
surface antigen CD146 recognized by the P1H12 antibody. These proteins have been shown to
be important in endothelial cell-cell interactions. These results led us to call these rhesus
embryonic stem cell-derived endothelial cells (RESDECs). Surprisingly, antibodies against the
VEGF receptors flk-1 and flt-1 did not bind these RESDECs, though binding to HUVEC cells
was also weak. However, an assay using biotinylated VEGF and secondary streptavidin-FITC
showed binding to the RESDEC (and HUVEC) cells. Specificity of this.binding was
demonstrated by blocking with an anti-VEGF antibody. This results suggests that either the anti-
human flk-1 and flt-1 antibodies did not cross-react with the rhesus-derived cells, or these cells
express a different VEGF receptor. RT-PCR studies showed expression of flk-1 mRNA in the
RESDECs, suggesting that the antibodies may not cross-react. Of course, it is possible that
protein derived from this mRNA is not properly expressed on the ceil surface.
[00042] The RESDEC cells do differ from HUVEC cells by lack of expression of CD31
and VE-cadherin, two surface antigens commonly, but not uniformly, found on the surface of
endothelial cells. RT-PCR studies confirm absence of mRNA expression of these genes.
However, some studies of mouse ES cell-derived endothelial cells also shows lack of CD31 and
VE-cadherin in certain endothelial cell populations. While CD31 and VE-cadherin can serve as
positive markers of endothelial cells, lack of expression does not preclude these being endothelial
cells.
[00043] Angiogenesis from RESDEC cells-
[00044] In vivo function of the RESDECs was first assessed by a Matrigel plug assay.
Here, RESDECs were imbedded in a sponge that was suspended into solidified Matrigel. This
Matrigel plug is then implanted subcutaneously in a severe combined immunodeficient (SCID)
mouse. After approximately 28 days, the mouse was injeceted intravenously with a FITC—
dextran solution, followed by plug removal and imaging. This demonstrated intense vascular
localization toward the RESDEC containing sponge, a chemotactic-like event typical of
endothelial cells. From the appearance of this vascularization, it is likely that this represents an
area of murine vessel angiogenesis in response to RESDEC-derived factors. Indeed, subsequent
analysis.of RESDEC supernatant by ELISA demonstrated a significant level of vascular
endothelial growth factor (VEGF) produced by these cells. However, basic fibroblast growth
factor (bFGF), another angiogenic protein often produced by endothelial cells, was not measured
in RESDEC culture supernatant.
[00045] To demonstrate neo-vessels produced by the RESDECs.. 0.5 -1.0 x 106 cells were
evenly suspended in a Matrigel plug implanted subcutaneously in a SCID mouse. Again, the
animals were injected with FITC-dextran, followed by plug removal and imaging. Here, larger
vascular structures were seen and subsequent histological examination of the plug showed
vascular formation by the RESDECs.
[00046] Next, to demonstrate the ability of RESDEC cells to contribute to active
angiogenesis within tumors in vivo another SCBD mouse model was used. Here, cells of the
mouse mammary carcinoma line C755 were injected subcutaneously either alone or co-injected
with an equivalent number of RESDECs. Tumor growth was measured at regular intervals and
tumors grew significantly faster when co-injected with RESDECs. Importantly, 106 RESDECs
injected alone did not lead to any measurable tumor growth, demonstrating these cells are not
directly tumorigenic. These tumors were highly vascular and immunohistochemical staining of
the tumors with anti-human specific antibodies (that cross-react to rhesus monkey but not mouse
cells) clearly demonstrate a contribution to the endothelium from the RESDEC cells. Staining of
a substantial number of endothelial cells was positive using anti-MHC class I or anti-vWF
antibodies in tumors co-injected with RESDEC cells, but endothelial cells in tumors derived from
C755 alone were negative for these antigens.
WE CLAIM:
1. A method for directing the differentiation of primate embryonic stem cells into
endothelial cells comprising the steps of
(a) culturing a culture of embryonic stem cells in a medium containing vascular
endothelial cell growth factor, basic fibroblast growth factor, insulin-like
growth factor and epidermal growth factor; and
(b) sub-culturing cells which have the morphology of endothelial cells.
2. A method as claimed in claim 1 wherein the primate cells are rhesus monkey cells.
3. A method as claimed in claim 1 wherein the primate cells are human cells.
4. A method as claimed in claim 1 wherein the medium for the culture of embryonic
stem cells into endothelial cells also includes mammalian serum.
A method is described to induce primate embryonic stem cells to differentiate into
a relatively homogenous population of endothelial cells. The ES derived endothelial cells
have the general morphological and cell surface marker characteristics of endothelial
cells. The ES derived endothelial cells also are capable of inducing and participating in
blood vessel formation (or vascularization) when transplanted into tissue in vivo.

Documents:

457-kolnp-2004-granted-abstract.pdf

457-kolnp-2004-granted-claims.pdf

457-kolnp-2004-granted-correspondence.pdf

457-kolnp-2004-granted-description (complete).pdf

457-kolnp-2004-granted-form 1.pdf

457-kolnp-2004-granted-form 3.pdf

457-kolnp-2004-granted-form 5.pdf

457-kolnp-2004-granted-gpa.pdf

457-kolnp-2004-granted-letter patent.pdf

457-kolnp-2004-granted-others.pdf

457-kolnp-2004-granted-reply to examination report.pdf

457-kolnp-2004-granted-specification.pdf


Patent Number 218574
Indian Patent Application Number 00457/KOLNP/2004
PG Journal Number 14/2008
Publication Date 04-Apr-2008
Grant Date 02-Apr-2008
Date of Filing 07-Apr-2004
Name of Patentee WISCONSIN ALUMNI RESEARCH FOUNDATION
Applicant Address 614, WALNUT STREET P.O. BOX 7365 MADISON WI53707-7365 USA.
Inventors:
# Inventor's Name Inventor's Address
1 KAUFMAN DANS N/A8391 HIDDEN PONDES ALCOVE, WOODBURY, MN 55125 USA.
2 LEWIS RACHEL 2943, KING JAMES WAY MADISON WI 53719 USA.
3 AUERBACH ROBERT 4905, FOXFIRE TRAIL MIDDLETON WI 53562 USA.
PCT International Classification Number C12N
PCT International Application Number PCT/US02/35278
PCT International Filing date 2002-11-01
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/335,332 2001-11-02 U.S.A.