Language selection

Search

Patent 2495824 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2495824
(54) English Title: EX-VIVO EXPANSION OF HEMATOPOIETIC STEM CELL POPULATIONS IN MONONUCLEAR CELL CULTURES
(54) French Title: EXPANSION EX VIVO DE POPULATIONS DE CELLULES SOUCHES HEMATOPOIETI QUES DANS DES CULTURES DE CELLULES MONONUCLEAIRES
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • C12N 5/0789 (2010.01)
  • C12N 5/078 (2010.01)
  • A01N 1/02 (2006.01)
  • A61K 35/28 (2015.01)
  • C12N 5/00 (2006.01)
  • C12N 5/10 (2006.01)
  • C12N 15/09 (2006.01)
  • C12N 15/87 (2006.01)
  • C12Q 1/02 (2006.01)
  • C12N 15/85 (2006.01)
(72) Inventors :
  • PELED, TONY (Israel)
  • TREVES, AVI (Israel)
  • ROSEN, OREN (Israel)
(73) Owners :
  • GAMIDA-CELL LTD. (Israel)
(71) Applicants :
  • GAMIDA-CELL LTD. (Israel)
(74) Agent: NORTON ROSE FULBRIGHT CANADA LLP/S.E.N.C.R.L., S.R.L.
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2003-08-17
(87) Open to Public Inspection: 2004-02-26
Examination requested: 2008-06-02
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/IL2003/000681
(87) International Publication Number: WO2004/016731
(85) National Entry: 2005-02-16

(30) Application Priority Data:
Application No. Country/Territory Date
60/404,137 United States of America 2002-08-19
60/404,145 United States of America 2002-08-19
152904 Israel 2002-11-17
PCT/IL03/00062 Israel 2003-01-23
PCT/IL03/00064 Israel 2003-01-26
60/452,545 United States of America 2003-03-07

Abstracts

English Abstract




Ex-vivo methods of expanding hematopoietic stem cells of hematopoietic
mononuclear cells that comprise a major fraction of hematopoietic committed
cells and a minor fraction of hematopoietic stem and progenitor cells,
expanded populations of hematopoietic stem cells obtained thereby and their
uses are disclosed


French Abstract

L'invention concerne des procédés ex vivo d'expansion de cellules souches hématopoïétiques de cellules mononucléaires hématopoïétiques qui comprennent une fraction majeure de cellules hématopoïétiques engagées et une fraction mineure de cellules souches hématopoïétiques et de cellules progénitrices hématopoïétiques, et des populations développées de cellules souches hématopoïétiques obtenues à partir de ces procédés ainsi que leurs utilisations.

Claims

Note: Claims are shown in the official language in which they were submitted.





97


WHAT IS CLAIMED IS:

1. A method of ex-vivo expanding a population of hematopoietic stem
cells, while at the same time, substantially inhibiting differentiation of the
hematopoietic stem cells ex-vivo, the method comprising providing
hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells, with ex-vivo
culture
conditions for ex-vivo cell proliferation and, at the same time, for reducing
an
expression and/or activity of CD38, thereby expanding a population of said
hematopoietic stem cells, while at the same time, substantially inhibiting
differentiation of said hematopoietic stem cells ex-vivo.

2. A method of hematopoietic cells transplantation or implantation
comprising:
(a) obtaining hematopoietic mononuclear cells which comprise a major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells from a donor;
(b) providing said hematopoietic mononuclear cells with ex-vivo culture
conditions for cell proliferation and, at the same time, for reducing an
expression
and/or activity of CD38, thereby expanding a population of said hematopoietic
stem
cells, while at the same time, substantially inhibiting differentiation of
said
hematopoietic stem cells ex-vivo; and
(c) transplanting or implanting said hematopoietic stem cells to a recipient.

3. The method of claim 2, wherein said donor and said recipient are a
single individual.

4. A method of genetically modifying hematopoietic stem cells with an
exogene comprising:

(a) obtaining hematopoietic mononuclear cells which comprise a major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells;


98

(b) providing said hematopoietic mononuclear cells with ex-vivo culture
conditions for cell proliferation and, at the same time, for reducing an
expression
and/or activity of CD38, thereby expanding a population of said hematopoietic
stem
cells, while at the same time, substantially inhibiting differentiation of
said
hematopoietic stem cells ex-vivo; and
(c) genetically modifying said hematopoietic stem cells with the exogene.

5. The method of claim 4, wherein genetically modifying is effected by a
vector which comprises the exogene.

6. The method of claim 5, wherein the vector is a viral vector or a nucleic
acid vector.

7. A method of adoptive immunotherapy comprising:
(a) obtaining hematopoietic mononuclear cells which comprise a major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells from a recipient;
(b) providing said hematopoietic mononuclear cells with ex-vivo culture
conditions for cell proliferation and, at the same time, for reducing an
expression
and/or activity of CD38, thereby expanding a population of said hematopoietic
stem
cells, while at the same time, substantially inhibiting differentiation of
said
hematopoietic stem cells; and
(c) transplanting said hematopoietic stem cells to the recipient.

8. A transplantable hematopoietic cell preparation comprising an
expanded population of hematopoietic stem cells propagated ex-vivo from
hematopoietic mononuclear cells which comprise, prior to expansion, a major
fraction
of hematopoietic committed cells and a minor fraction of hematopoietic stem
and
progenitor cells, in the presence of an effective amount of an agent, said
agent
reducing an expression and/or activity of CD38, while at the same time,
substantially
inhibiting differentiation of said hematopoietic stem cells, and a
pharmaceutically
acceptable carrier.




99

9. The method of any of claims 1, 2, 4, and 7, wherein said hematopoietic
mononuclear cells are derived from a source selected from the group consisting
of
bone marrow, peripheral blood and neonatal umbilical cord blood.

10. The method of any of claims 1, 2, 4 and 7, wherein providing said
hematopoietic mononuclear cells with said conditions for ex-vivo cell
proliferation
comprises providing said hematopoietic mononuclear cells with nutrients and
with
cytokines.

11. The method of claim 10, wherein said cytokines are early acting
cytokines.

12. The method of claim 11, wherein said early acting cytokines are
selected from the group consisting of stem cell factor, FLT3 ligand,
interleukin-1,
interleukin-2, interleukin-3, interleukin-6, interleukin-10, interleukin-12,
tumor
necrosis factor-a and thrombopoietin.

13. The method of claim 10, wherein said cytokines are late acting
cytokines.

14. The method of claim 13, wherein said late acting cytokines are selected
from the group consisting of granulocyte colony stimulating factor,
granulocyte/macrophage colony stimulating factor, erythropoietin, FGF, EGF,
NGF,
VEGF, LIF, Hepatocyte growth factor and macrophage colony stimulating factor.

15. The method of any of claims 1, 2, 4 and 7, wherein providing said
hematopoietic mononuclear cells with ex-vivo culture conditions for reducing
said
expression and/or said activity of CD38 is by providing said hematopoietic
mononuclear cells with an agent that downregulates CD38 expression.

16. The transplantable hematopoietic cell preparation of claim 8, wherein
said agent is an agent that downregulates CD38 expression.




100

17. The method of any of claims 15 and 16, wherein said agent that
downregulates CD38 expression is selected from the group consisting of a
retinoic
acid receptor antagonist, a retinoid X receptor antagonist and a Vitamin D
receptor
antagonist.

18. The method of any of claims 15 and 16, wherein said agent that
downregulates CD38 expression is an antagonist for reducing a capacity of said
hematopoietic mononuclear cells in responding to retinoic acid, retinoid
and/or
Vitamin D.

19. The method of any of claims 15 and 16, wherein said agent that
downregulates CD38 expression is a polynucleotide.

20. The method of claim 19, wherein said polynucleotide encodes an anti
CD38, an anti retinoic acid receptor, an anti retinoid X receptor or an anti
Vitamin D
receptor intracellular antibody.

21. The method of claim 19, wherein said polynucleotide encodes an anti
CD38, an anti retinoic acid receptor, an anti retinoid X receptor or an anti
Vitamin D
receptor antibody.

22. The method of claim 19, wherein said polynucleotide is a small
interfering polynucleotide molecule directed to cause intracellular CD38,
retinoic
acid receptor, retinoid X receptor or Vitamin D receptor mRNA degradation.

23. The method of claim 22, wherein said small interfering
polynucleotide molecule is selected from the group consisting of an RNAi
molecule,
an anti-sense molecule, a rybozyme molecule and a DNAzyme molecule.

24. The method of any of claims 15 and 16, wherein said agent that
downregulates CD38 expression is an agent that downregulates PI 3-kinase
expression.




101

25. The method of claim 24, wherein said agent that downregulates PI 3-
kinase expression is a polynucleotide.
26. The method of claim 24, wherein agent that downregulates PI 3-
kinase expression is an intracellular antibody.
27. The method of claim 25, wherein said polynucleotide is a small
interfering polynucleotide molecule directed to cause intracellular PI 3-
kinase
mRNA or gene degradation.
28. The method of claim 27, wherein said small interfering
polynucleotide molecule is selected from the group consisting of an RNAi
molecule,
an anti-sense molecule, a rybozyme molecule and a DNAzyme molecule.
29. The method of any of claims 15 and 16, wherein said agent that
downregulates CD38 expression is an agent that inhibits PI 3-kinase activity.
30. The method of claim 30, wherein said agent that inhibits PI 3-kinase
activity is selected from the group consisting of wortmannin and LY294002
31. The method of any of claims 1, 2, 4 and 7, wherein providing said
hematopoietic mononuclear cells with ex-vivo culture conditions for reducing
said
expression and/or said activity of CD38 is by providing said hematopoietic
mononuclear cells with an agent that inhibits CD38 activity.
32. The transplantable hematopoietic cell preparation of claim 8, wherein
said agent is an agent that inhibits CD38 activity.
33. The method of any of claims 31 and 32, wherein said agent that
inhibits CD38 activity is nicotinamide, a nicotinamide analog, a nicotinamide
or a
nicotinamide analog derivative or a nicotinamide or a nicotinamide analog
metabolite.



102


34. The method of claim 33, wherein said nicotinamide analog is selected
from the group consisting of benzamide, nicotinethioamide, nicotinic acid and
.alpha.-
amino-3-indolepropionic acid.
35. The method of any of claims 1, 2, 4 and 7, wherein providing said
hematopoietic mononuclear cells with ex-vivo culture conditions for reducing
said
expression and/or said activity of CD38 is by providing said hematopoietic
mononuclear cells with an agent that inhibits PI 3-kinase activity.
36. The transplantable hematopoietic cell preparation of claims 8,
wherein said agent is an agent that inhibits PI 3-kinase activity.
37. The method of any of claims 35 and 36, wherein said agent that
inhibits PI 3-kinase activity is selected from the group consisting of
wortmannin and
LY294002.
38. A method of ex-vivo expanding a population of hematopoietic stem
cells, while at the same time, substantially inhibiting differentiation of the
hematopoietic stem cells ex-vivo, the method comprising providing
hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells, with ex-vivo
culture
conditions for ex-vivo cell proliferation and, at the same time, for reducing
a capacity
of said hematopoietic mononuclear cells in responding to retinoic acid,
retinoids
and/or Vitamin D, thereby expanding a population of said hematopoietic stem
cells
while at the same time, substantially inhibiting differentiation of said
hematopoietic
stem cells ex-vivo.
39. A method of hematopoietic cells transplantation or implantation
comprising:
(a) obtaining hematopoietic mononuclear cells which comprise a major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells from a donor;




103


(b) providing said hematopoietic mononuclear cells with ex-vivo culture
conditions for cell proliferation and, at the same time, for reducing a
capacity of said
hematopoietic mononuclear cells in responding to retinoic acid, retinoids
and/or
Vitamin D, thereby expanding a population of said hematopoietic stem cells,
while at
the same time, substantially inhibiting differentiation of said hematopoietic
stem cells
ex-vivo; and
(c) transplanting or implanting said hematopoietic stem cells to a recipient.
40. The method of claim 39, wherein said donor and said recipient are a
single individual.
41. A method of genetically modifying hematopoietic stem cells with an
exogene comprising:
(a) obtaining hematopoietic mononuclear cells which comprise a major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells;
(b) providing said hematopoietic mononuclear cells with ex-vivo culture
conditions for cell proliferation and, at the same time, for reducing a
capacity of said
hematopoietic mononuclear cells in responding to retinoic acid, retinoids
and/or
Vitamin D, thereby expanding a population of said hematopoietic stem cells,
while at
the same time, substantially inhibiting differentiation of said hematopoietic
stem cells
ex-vivo; and
(c) genetically modifying said hematopoietic stem cells with the exogene.
42. The method of claim 41, wherein genetically modifying is effected by
a vector which comprises the exogene.
43. The method of claim 42, wherein the vector is a viral vector or a
nucleic acid vector.
44. A method of adoptive immunotherapy comprising:




104


(a) obtaining hematopoietic mononuclear cells which comprise a major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells from a recipient;
(b) providing said hematopoietic mononuclear cells with ex-vivo culture
conditions for cell proliferation and, at the same time, for reducing a
capacity of said
hematopoietic mononuclear cells in responding to retinoic acid, retinoids
and/or
Vitamin D, thereby expanding a population of said hematopoietic stem cells,
while at
the same time, substantially inhibiting differentiation of said hematopoietic
stem cells;
and
(c) transplanting said hematopoietic stem cells to the recipient.
45. A transplantable hematopoietic cell preparation comprising an
expanded population of hematopoietic stem cells propagated ex-vivo from
hematopoietic mononuclear cells which comprise, prior to expansion, a major
fraction
of hematopoietic committed cells and a minor fraction of hematopoietic stem
and
progenitor cells in the presence of an effective amount of an agent, said
agent
reducing a capacity of said hematopoietic mononuclear cells in responding to
retinoic
acid, retinoids and/or Vitamin D, while at the same time, substantially
inhibiting
differentiation of said hematopoietic stem cells, and a pharmaceutically
acceptable
carrier.
46. The method and/or the transplantable hematopoietic cell preparation of
any of claims 38, 39, 41, 44 and 45, wherein said hematopoietic mononuclear
cells are
derived from a source selected from the group consisting of bone marrow,
peripheral
blood and neonatal umbilical cord blood.
47. The method of any of claims 38, 39, and 44, wherein providing said
hematopoietic mononuclear cells with said conditions for ex-vivo cell
proliferation
comprises providing said hematopoietic mononuclear cells with nutrients and
with
cytokines.
48. The method of claim 47, wherein said cytokines are early acting
cytokines.


105
49. The method of claim 48, wherein said early acting cytokines are
selected from the group consisting of stem cell factor, FLT3 ligand,
interleukin-l,
interleukin-2, interleukin-3, interleukin-6, interleukin-10, interleukin-12,
tumor
necrosis factor-a and thrombopoietin.
50. The method of claim 47, wherein said cytokines are late acting
cytokines.
51. The method of claim 50, wherein said late acting cytokines are
selected from the group consisting of granulocyte colony stimulating factor,
granulocyte/macrophage colony stimulating factor, erythropoietin, FGF, EGF,
NGF,
VEGF, LIF, Hepatocyte growth factor and macrophage colony stimulating factor.
52. The method and/or the transplantable hematopoietic cell preparation of
any of claims 38, 39, 41, 44 and 45, wherein reducing said capacity of said
hematopoietic mononuclear cells in responding to retinoic acid, retinoids
and/or
Vitamin D is reversible.
53. The method and/or the transplantable hematopoietic cell preparation of
any of claims 38, 39, 41, 44 and 45, wherein reducing said capacity of said
hematopoietic mononuclear cells in responding to retinoic acid, retinoids
and/or
Vitamin D is by ex-vivo culturing said hematopoietic mononuclear cells in a
presence
of an effective amount of at least one retinoic acid receptor antagonist, at
least one
retinoid X receptor antagonist and/or at least one Vitamin D receptor
antagonist.
54. The method of claim 53, wherein reducing said capacity of said
hematopoietic mononuclear cells in responding to retinoic acid, retinoids
and/or
Vitamin D is by ex-vivo culturing said hematopoietic mononuclear cells in a
presence
of an effective amount of at least one retinoic acid receptor antagonist, at
least one
retinoid X receptor antagonist and/or at least one Vitamin D receptor
antagonist, for a
time period of 0.1-50 % of an entire ex-vivo culturing period of said
hematopoietic
mononuclear cells.


106

55. The transplantable hematopoietic cell preparation of claim 45, wherein
said agent is selected from the group consisting of retinoic acid receptor
antagonist,
retinoid X receptor antagonist and/or Vitamin D receptor antagonist.
56. The method and/or the transplantable hematopoietic cell preparation of
any of claims 53, 54 and 55, wherein said retinoic acid receptor antagonist is
selected
from the group consisting of:
AGN 194310; AGN 193109; 3-(4-Methoxy-phenylsulfanyl)-3-methyl-butyric acid;
6-Methoxy-2,2-dimethvl-thiochroman-4-one,2,2-Dimethyl-4-oxo-thiochroman-6-
yltrifluoromethane-sulfonate; Ethyl 4-((2,2 dimethyl-4-oxo-thiochroman-6-
yl)ethynyl)-benzoate; Ethyl 4-((2,2-dimethy 1-4-triflouromethanensulfonyloxy -
(2H)-
thiochromen-6-yl)ethynyl)-benzoate(41); Thiochromen-6-yl]-ethynyl]-
benzoate(yl);
(p-[(E)-2-[3'4'-Dihydro-4,4'-dimethyl-7'-(heptyloxy)-2'H-1-benzothiopyran-
6'yl]
propenyl] benzoic acid 1'1'-dioxide; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
butoxyphenyl)-
3-methyl]-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
propoxyphenyl)-
3-methyl]-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
pentoxyphenyl)-
3-methyl]-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
hexoxyphenyl)-3-
methyl]-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
heptoxyphenyl)-3-
methyl]-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
octoxyphenyl)-3-
methyl]-octa-2,4,6-trienoic acid; (2E,4E,6E)-7-[3-t-butyl-5-(1-phenyl-vinyl)-
phenyl]-
3-methyl-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-{[4,5-3
H2]-n-pentoxy}phenyl)-3-methyl]-octa-2,4,6-trienoic acid; (2E,4E)-
(1RS,2RS)-
5-[2-(3,5-di-tert.butyl-2-ethoxy-phenyl)-cyclopropyl]-3-methyl-penta-2,4-
dienoic acid
ethyl ester; (2E,4E)-(1RS,2RS)-5-[2-(3,5-di-tert.butyl-2-ethoxy-phenyl)-
cyclopropyl]-
3-methyl-penta-2,4-dienoic acid; (2E,4E)-(1RS,2RS)-5-[2-(3,5-di-tert.butyl-2-
butoxy-phenyl)-cyclopropyl]-3-methyl-penta-2,4-dienoic(2E,4E,6Z)-7-[3,5-di-
acid;


tert.butyl-2-ethoxyphenyl]3-methyl-2,4,6-octatrienoic(2E,4E,6Z)-7-[3,5-di-
acid;


tert.butyl-2-butyloxyphenyl]-3-methyl-2,4,6-octatrienoic4-(5,6,7,8-tetrahydro-
acid;


5,5,8,8-tetramethyl-2-naphthalene-carboxamido)(2E,4E)-3-methyl-5-
benzoic acid;


[(1S,2S)-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydro-naphthalen-2-yl)-
cyclopropyl]-
penta-2,4-dienoic acid; p-[(E)-2-[3',4'-Dihydro-4',4'-dimethyl-7'-(heptyloxy)-
2'H-1-
benzothiopyran-6'-yl]propenyl]benzoic acid; 1',1'-dioxide, 4-(7,7,10,10-
Tetramethyl-
1-pyridin-3-ylmethyl-4,5,7,8,9,10-hexahydro-1H-naphto[2,3-g]indol-3-yl)-
benzoic


107
acid; (2E,4E,6Z)-7-[3,5-di-tert.butyl-2-methoxyphenyl]-3-methyl-2,4,6-
octatrienoic
acid; (2E,4E,6Z)-7-[3,5-di-tert.butyl-2-ethoxyphenyl]-3-methyl-2,4,6-
octatrienoic
acid; (2E,4E,6Z)-7-[3,5-di-tert.butyl-2-hexyloxyphenyl]-3-methyl-2,4,6-
octatrienoic
acid; (2E,4E,6Z)-7-[3,5-di-tert.butyl-2-octyloxyphenyl]-3-methyl-2,4,6-
octatrienoic
acid; and (2E,4E)-(1RS,2RS)-5-[2-(3,5-di-tert-butyl-2-butoxy-phenyl)-
cyclopropyl]-
3-methyl-penta-2,4-dienoic acid, (2E,4E,6Z)-7-(3-n-propoxy-5,6,7,8-tetrahydro-
5,5,8,8-tetramethylnaphthalene-2-yl)-3-methylocta-2,4,6-trienoic acid, 4-(5H-
2,3(2,5
dimethyl-2,5-hexano)-5-n-propyldibenzo[b,e][1,4]diazepin-11-yl)benzoic acid, 4-

(SH-2,3-(2,5-dimethyl-2,5-hexano)-5methyl-8-nitrodibenzo[b,e][1,4]diazepin-11-
yl)benzoic acid, 4-{[4-(4-Ethylphenyl)2,2-dimethyl-(2H)-thiochromen-6-
yl]ethynyl}benzoic acid, 4-[4-2methyl-1,2-dicarba-closo-dodecaboran-1-yl-
phenylcarbamoyl]benzoic acid, 4-[4,5,7,8,9,10-hexahydro-7,7,10,10-tetramethyl-
1-(3-
pyridylmethyl)-anthra[1,2-b]pyrrol-3-yl]benzoic acid, (3-pyridylmethyl)-]5-
thiaanthra[2,1-b]pyrrol-3-yl)benzoic acid, and (3-pyridylmethyl)-anthra[2m1-
d]pyrazol-3-yl]benzoic acid.
57. The method and/or the transplantable hematopoietic cell preparation of
any of claims 53, 54 and 55, wherein said retinoid X receptor antagonist is
selected
from the group consisting of:
LGN100572, LGN100574, 1-(3-hydroxy-5,6,7,8-tetrahydro-5,5,8,8-
tetramethylnaphthalene-2-yl)ethanone, 1-(3-propoxy-5,6,7,8-tetrahydro-5,5,8,8-
tetramethylnaphthalene-2-yl)ethanone, 3-(3-propoxy-5,6,7,8-tetrahydro-5,5,8,8-
tetramethylnaphthalene-2-yl)but-2-enenitrile, 3-(3-propoxy-5,6,7,8-tetrahydro-
5,5,8,8-tetramethylnaphthalene-2-yl)but-2-enal, (2E,4E,6E)-7-3[-propoxy-
5,6,7,8-
tetrahydro 5,5,8,8-tetramethyl-2-naphthalene-2-yl]-3-methylocta-2,4,6-trienoic
acid,
4-[3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)carbonyl] benzoic acid,
4-[1-
(3,5, 5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)ethenyl] benzoic acid, 4-

[1(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)cyclopropyl] benzoic
acid, 4-
[1-(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)ethenyl] benzenete
trazole,
2-[1-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydro-2-naphthyl) ethenyl]pyridine-5-
carboxylic acid, 2-[1-(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-
naphthyl)ethyl]pyridine-5-carboxylic acid, ethyl-2-[1-(3,5,5,8, 8-pentamethyl-
5,6,7,8-
tetrahydro-2-naphthyl)ethenyl]pyridine-5-carboxylate, 5-[1-3,5,5,8,8-
pentamethyl-



108

5,6,7,8-tetrahydro-2-naphthyl)ethenyl]pyridine-2-carboxylic acid, 2-[1-
(3,5,5,8,8-
pentamethyl-5,6,7,8-tetrahydro-2-naphthyl) cyclopropyl]pyridine-5-carboxylic
acid,
methyl 2-[ 1-(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-
naphthyl)cyclopropyl]pyridine-5-carboxylate, 4-[1-(3,5, 5,8,8-pentamethyl-
5,6,7,8-
tetrahydro-2-naphthyl)ethenyl]-N-(4-hydroxyphenyl) benzamide, 2-[1-(3,5,5,8,8-
Pentamethyl-5,6,7,8-tetrahydro-2-naphthyl) ethenyl] pyridine-5-carboxylic
acid, 2-[1-
(3,5,5,8,8-Pentamethyl-5, 6,7,8-tetrahydro-2-naphthyl)cyclopropyl]pyridine-5-
carboxylic acid, 4-[(3,5, 5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-
naphthyl)carbonyl]benzoic acid butyloxime, 4-[(3,5,5,8,8-pentamethyl-5,6,7,8-
tetrahydro-2-naphthyl) carbonyl]benzoic acid propyloxime, 4-[(3,5,5,8,8-
pentamethyl-5,6,7,8-terrahydro-2-naphthyl)carbonyl]benzoic acid cyanoimine, 4-
[(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)carbonyl]benzoic acid
allyloxime, 4-[(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-
naphthyl)carbonyl]benzoic
acid 4-(3-methylbut-2-enoic acid)oxime, and 4-[(3,5,5,8,8-pentamethyl-5,6,7,8-
tetrahydro-2-naphthyl)carbonyl]benzoic acid 1-aminoethyloxime, (2E,4E,6Z)-7-(3-
n-
propoxy-5,6,7,8-tetrahydro-5,5,8,8-tetramethylnaphthalene-2-yl)-3-methylocta-
2,4,6-
trienoic acid, 4-(5H-2,3(2,5 dimethyl-2,5-hexano)-5-n-
propyldibenzo[b,e][1,4]diazepin-11-yl)benzoic acid, and 4-(5H-2,3-(2,5-
dimethyl-
2,5-hexano)-5methyl-8-nitrodibenzo[b,e][1,4]diazepin-11-yl)benzoic acid.
58. The method and/or the transplantable hematopoietic cell preparation of
any of claims 53, 54 and 55, wherein said Vitamin D receptor antagonist is
selected
from the group consisting of: 1 alpha, 25-(OH)-D3-26,23 lactone; 1alpha, 25-
dihydroxyvitamin D (3); the 25-carboxylic ester ZK159222; (23S)- 25-dehydro-1
alpha-OH-D (3); (23R)-25-dehydro-1 alpha-OH-D (3); 1 beta, 25 (OH)Z D3; 1
beta,
25(OH)2-3-epi-D3; (23S) 25-dehydro-1 alpha(OH) D3-26,23-lactone; (23R) 25-
dehydro-1 alpha(OH)D3-26,23-lactone and Butyl-(5Z,7E,22E-(1S,7E,22E-
(1 S,3R,24R)-1,3,24-trihydroxy-26,27-cyclo-9,10-secocholesta-5,7,10(19),22-
tetraene-
25-carboxylate).
59. A method of ex-vivo expanding a population of hematopoietic stem
cells, while at the same time, substantially inhibiting differentiation of the
hematopoietic stem cells ex-vivo, the method comprising providing
hematopoietic



109
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells, with ex-vivo
culture
conditions for ex-vivo cell proliferation and, at the same time, for reducing
a capacity
of said hematopoietic mononuclear cells in responding to signaling pathways
involving the retinoic acid receptor, the retinoid X receptor and/or the
Vitamin D
receptor, thereby expanding a population of said hematopoietic stem cells
while at the
same time, substantially inhibiting differentiation of said hematopoietic stem
cells ex-
vivo.
60. A method of hematopoietic cells transplantation or implantation
comprising:
(a) obtaining hematopoietic mononuclear cells which comprise a major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells from a donor;
(b) providing said hematopoietic mononuclear cells with ex-vivo culture
conditions for cell proliferation and, at the same time, for reducing a
capacity of said
hematopoietic mononuclear cells in responding to signaling pathways involving
the
retinoic acid receptor, the retinoid X receptor and/or the Vitamin D receptor,
thereby
expanding a population of said hematopoietic stem cells, while at the same
time,
substantially inhibiting differentiation of said hematopoietic stem cells ex-
vivo; and
(c) transplanting or implanting said hematopoietic stem cells to a recipient.
61. The method of claim 60, wherein said donor and said recipient are a
single individual.
62. A method of genetically modifying hematopoietic stem cells with an
exogene comprising:
(a) obtaining hematopoietic mononuclear cells which comprise a major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells;
(b) providing said hematopoietic mononuclear cells with ex-vivo culture
conditions for cell proliferation and, at the same time, for reducing a
capacity of said
hematopoietic mononuclear cells in responding to signaling pathways involving
the


110

retinoic acid receptor, the retinoid X receptor and/or the Vitamin D receptor,
thereby
expanding a population of said hematopoietic stem cells, while at the same
time,
substantially inhibiting differentiation of said hematopoietic stem cells ex-
vivo; and
(c) genetically modifying said hematopoietic stem cells with the exogene.
63. The method of claim 62, wherein genetically modifying is effected by
a vector which comprises the exogene.
64. The method of claim 63, wherein the vector is a viral vector or a
nucleic acid vector.
65. A method of adoptive immunotherapy comprising:
(a) obtaining hematopoietic mononuclear cells which comprise a major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells from a recipient;
(b) providing said hematopoietic mononuclear cells with ex-vivo culture
conditions for cell proliferation and, at the same time, for reducing a
capacity of said
hematopoietic mononuclear cells in responding to signaling pathways involving
the
retinoic acid receptor, the retinoid X receptor and/or the Vitamin D receptor,
thereby
expanding a population of said hematopoietic stem cells, while at the same
time,
substantially inhibiting differentiation of said hematopoietic stem cells; and
(c) transplanting said hematopoietic stem cells to the recipient.
66. A transplantable hematopoietic cell preparation comprising an
expanded population of hematopoietic stem cells propagated ex-vivo from
hematopoietic mononuclear cells which comprise, prior to expansion, a major
fraction
of hematopoietic committed cells and a minor fraction of hematopoietic stem
and
progenitor cells in the presence of an effective amount of an agent, said
agent
reducing a capacity of said hematopoietic mononuclear cells in responding to
signaling pathways involving the retinoic acid receptor, the retinoid X
receptor and/or
the Vitamin D receptor, while at the same time, substantially inhibiting
differentiation
of said hematopoietic stem cells, and a pharmaceutically acceptable carrier.



111

67. The method and/or the transplantable hematopoietic cell preparation of
any of claims 59, 60, 62, 65 and 66, wherein said hematopoietic mononuclear
cells are
derived from a source selected from the group consisting of bone marrow,
peripheral
blood and neonatal umbilical cord blood.
68. The method of any of claims 59, 60, and 65, wherein providing said
hematopoietic mononuclear cells with said conditions for ex-vivo cell
proliferation
comprises providing said hematopoietic mononuclear cells with nutrients and
with
cytokines.
69. The method of claim 68, wherein said cytokines are early acting
cytokines.
70. The method of claim 69, wherein said early acting cytokines are
selected from the group consisting of stem cell factor, FLT3 ligand,
interleukin-1,
interleukin-2, interleukin-3, interleukin-6, interleukin-10, interleukin-12,
tumor
necrosis factor-a and thrombopoietin.
71. The method of claim 68, wherein said cytokines are late acting
cytokines.
72. The method of claim 71, wherein said late acting cytokines are
selected from the group consisting of granulocyte colony stimulating factor,
granulocyte/macrophage colony stimulating factor, erythropoietin, FGF, EGF,
NGF,
VEGF, LIF, Hepatocyte growth factor and macrophage colony stimulating factor.
73. The method and/or the transplantable hematopoietic cell preparation of
any of claims 59, 60, 62, 65 and 66, wherein reducing said capacity of said
hematopoietic mononuclear cells in responding to signaling pathways involving
the
retinoic acid receptor, the retinoid X receptor and/or the Vitamin D receptor
is
reversible.



112
74. The method and/or the transplantable hematopoietic cell preparation of
any of claims 59, 60, 62, 65 and 66, wherein reducing said capacity of said
hematopoietic mononuclear cells in responding to signaling pathways involving
the
retinoic acid receptor, the retinoid X receptor and/or the Vitamin D receptor
is by ex-
vivo culturing . said hematopoietic mononuclear cells fraction in a presence
of an
effective amount of at least one retinoic acid receptor antagonist, at least
one retinoid
X receptor antagonist and/or at least one Vitamin D receptor antagonist.
75. The method of claim 74, wherein reducing said capacity of said
hematopoietic mononuclear cells in responding to signaling pathways involving
the
retinoic acid receptor, the retinoid X receptor and/or the Vitamin D receptor
is by ex-
vivo culturing said hematopoietic mononuclear cells in a presence of an
effective
amount of at least one retinoic acid receptor antagonist, at least one
retinoid X
receptor antagonist and/or at least one Vitamin D receptor antagonist, for a
time
period of 0.1-50 % of an entire ex-vivo culturing period of said hematopoietic
mononuclear cells.
76. The transplantable hematopoietic cell preparation of claim 66, wherein
said agent is selected from the group consisting of retinoic acid receptor
antagonist,
retinoid X receptor antagonist and/or Vitamin D receptor antagonist.
77. The method and/or the transplantable hematopoietic cell preparation of
any of claims 74, 75 and 76, wherein said retinoic acid receptor antagonist is
selected
from the group consisting of:
AGN 194310; AGN 193109; 3-(4-Methoxy-phenylsulfanyl)-3-methyl-butyric acid;
6-Methoxy-2,2-dimethvl-thiochroman-4-one,2,2-Dimethyl-4-oxo-thiochroman-6-
yltrifluoromethane-sulfonate; Ethyl 4-((2,2 dimethyl-4-oxo-thiochroman-6-
yl)ethynyl)-benzoate; Ethyl 4-((2,2-dimethy 1-4-triflouromethanensulfonyloxy -
(2H)-
thiochromen-6-yl)ethynyl)-benzoate(41); Thiochromen-6-yl]-ethynyl]-
benzoate(yl);
(p-[(E)-2-[3'4'-Dihydro-4,4'-dimethyl-7'-(heptyloxy)-2'H-1-benzothiopyran-
6'yl]
propenyl] benzoic acid 1'1'-dioxide; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
butoxyphenyl)-
3-methyl]-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
propoxyphenyl)-
3-methyl]-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
pentoxyphenyl)-


113

3-methyl]-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
hexoxyphenyl)-3-
methyl]-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
heptoxyphenyl)-3-
methyl]-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
octoxyphenyl)-3-
methyl]-octa-2,4,6-trienoic acid; (2E,4E,6E)-7-[3-t-butyl-5-(1-phenyl-vinyl)-
phenyl]-
3-methyl-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-{[4,5-3
H2]-n-pentoxy}phenyl)-3-methyl]-octa-2,4,6-trienoic acid; (2E,4E)-
(1RS,2RS)-
5-[2-(3,5-di-tert.butyl-2-ethoxy-phenyl)-cyclopropyl]-3-methyl-penta-2,4-
dienoic acid
ethyl ester; (2E,4E)-(1RS,2RS)-S-[2-(3,5-di-tert.butyl-2-ethoxy-phenyl)-
cyclopropyl]-
3-methyl-penta-2,4-dienoic acid; (2E,4E)-(1RS,2RS)-5-[2-(3,5-di-tert.butyl-2-
butoxy-phenyl)-cyclopropyl]-3-methyl-penta-2,4-dienoic acid; (2E,4E,6Z)-7-[3,5-
di-
tert.butyl-2-ethoxyphenyl]3-methyl-2,4,6-octatrienoic acid; (2E,4E,6Z)-7-[3,5-
di-
tert.butyl-2-butyloxyphenyl]-3-methyl-2,4,6-octatrienoic acid; 4-(5,6,7,8-
tetrahydro-
5,5,8,8-tetramethyl-2-naphthalene-carboxamido) benzoic acid; (2E,4E)-3-methyl-
5-
[( 1 S,2S)-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydro-naphthalen-2-yl)-
cyclopropyl]-
penta-2,4-dienoic acid; p-[(E)-2-[3',4'-Dihydro-4',4'-dimethyl-7'-(heptyloxy)-
2'H-1-
benzothiopyran-6'-yl]propenyl]benzoic acid; 1',1'-dioxide, 4-(7,7,10,10-
Tetramethyl-
1-pyridin-3-ylmethyl-4,5,7,8,9,10-hexahydro-1 H-naphto[2,3-g]indol-3-yl)-
benzoic
acid; (2E,4E,6Z)-7-[3,5-di-tert.butyl-2-methoxyphenyl]-3-methyl-2,4,6-
octatrienoic
acid; (2E,4E,6Z)-7-[3,5-di-tert.butyl-2-ethoxyphenyl]-3-methyl-2,4,6-
octatrienoic
acid; (2E,4E,6Z)-7-[3,5-di-tert.butyl-2-hexyloxyphenyl]-3-methyl-2,4,6-
octatrienoic
acid; (2E,4E,6Z)-7-[3,5-di-tert.butyl-2-octyloxyphenyl]-3-methyl-2,4,6-
octatrienoic
acid; and (2E,4E)-(1RS,2RS)-5-[2-(3,5-di-tert-butyl-2-butoxy-phenyl)-
cyclopropyl]-
3-methyl-penta-2,4-dienoic acid, (2E,4E,6Z)-7-(3-n-propoxy-5,6,7,8-tetrahydro-
5,5,8,8-tetramethylnaphthalene-2-yl)-3-methylocta-2,4,6-trienoic acid, 4-(5H-
2,3(2,5
dimethyl-2,5-hexano)-5-n-propyldibenzo[b,e][1,4]diazepin-11-yl)benzoic acid, 4-

(5H-2,3-(2,5-dimethyl-2,5-hexano)-5methyl-8-nitrodibenzo[b,e][ 1,4]diazepin-11-

yl)benzoic acid, 4-{[4-(4-Ethylphenyl)2,2-dimethyl-(2H)-thiochromen-6-
yl]ethynyl}benzoic acid, 4-[4-2methyl-1,2-dicarba-closo-dodecaboran-1-yl-
phenylcarbamoyl]benzoic acid, 4-[4,5,7,8,9,10-hexahydro-7,7,10,10-tetramethyl-
1-(3-
pyridylmethyl)-anthra[1,2-b]pyrrol-3-yl]benzoic acid, (3-pyridylmethyl)-]5-
thiaanthra[2,1-b]pyrrol-3-yl)benzoic acid, and (3-pyridylmethyl)-anthra[2m1-
d]pyrazol-3-yl]benzoic acid.




114


78. The method and/or the transplantable hematopoietic cell preparation of
any of claims 74, 75 and 76, wherein said retinoid X receptor antagonist is
selected
from the group consisting o~
LGN100572, LGN100574, 1-(3-hydroxy-5,6,7,8-tetrahydro-5,5,8,8-
tetramethylnaphthalene-2-yl)ethanone, 1-(3-propoxy-5,6,7,8-tetrahydro-5,5,8,8-
tetramethylnaphthalene-2-yl)ethanone, 3-(3-propoxy-5,6,7,8-tetrahydro-5,5,8,8-
tetramethylnaphthalene-2-yl)but-2-enenitrile, 3-(3-propoxy-5,6,7,8-tetrahydro-
5,5,8,8-tetramethylnaphthalene-2-yl)but-2-enal, (2E,4E,6E)-7-3[-propoxy-
5,6,7,8-
tetrahydro 5,5,8,8-tetramethyl-2-naphthalene-2-yl]-3-methylocta-2,4,6-trienoic
acid,
4-[3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)carbonyl] benzoic acid,
4-[1-
(3,5, 5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)ethenyl] benzoic acid, 4-

[1(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)cyclopropyl] benzoic
acid, 4-
[1-(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)ethenyl] benzenete
trazole,
2-(1-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydro-2-naphthyl) ethenyl]pyridine-5-
carboxylic acid, 2-[1-(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-
naphthyl)ethyl]pyridine-5-carboxylic acid, ethyl-2-[1-(3,5,5,8, 8-pentamethyl-
5,6,7,8-
tetrahydro-2-naphthyl)ethenyl]pyridine-5-carboxylate, 5-[1-3,5,5,8,8-
pentamethyl-
5,6,7,8-tetrahydro-2-naphthyl)ethenyl]pyridine-2-carboxylic acid, 2-[1-
(3,5,5,8,8-
pentamethyl-5,6,7,8-tetrahydro-2-naphthyl) cyclopropyl]pyridine-5-carboxylic
acid,
methyl 2-[ 1-(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-
naphthyl)cyclopropyl]pyridine-5-carboxylate, 4-[1-(3,5, 5,8,8-pentamethyl-
5,6,7,8-
tetrahydro-2-naphthyl)ethenyl]-N-(4-hydroxyphenyl) benzamide, 2-[1-(3,5,5,8,8-
Pentamethyl-5,6,7,8-tetrahydro-2-naphthyl) ethenyl] pyridine-5-carboxylic
acid, 2-[1-
(3,5,5,8,8-Pentamethyl-5, 6,7,8-tetrahydro-2-naphthyl)cyclopropyl]pyridine-S-
carboxylic acid, 4-[(3,5, 5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-
naphthyl)carbonyl]benzoic acid butyloxime, 4-[(3,5,5,8,8-pentamethyl-5,6,7,8-
tetrahydro-2-naphthyl) carbonyl]benzoic acid propyloxime, 4-[(3,5,5,8,8-
pentamethyl-5,6,7,8-terrahydro-2-naphthyl)carbonyl]benzoic acid cyanoimine, 4-
[(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)carbonyl]benzoic acid
allyloxime, 4-[(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-
naphthyl)carbonyl]benzoic
acid 4-(3-methylbut-2-enoic acid)oxime, and 4-[(3,5,5,8,8-pentamethyl-5,6,7,8-
tetrahydro-2-naphthyl)carbonyl]benzoic acid 1-aminoethyloxime, (2E,4E,6Z)-7-(3-
n-
propoxy-5,6,7,8-tetrahydro-5,5,8,8-tetramethylnaphthalene-2-yl)-3-methylocta-
2,4,6-




115


trienoic acid, 4-(5H-2,3(2,5 dimethyl-2,5-hexano)-5-n-
propyldibenzo[b,e][1,4]diazepin-11-yl)benzoic acid, and 4-(5H-2,3-(2,5-
dimethyl-
2,5-hexano)-Smethyl-8-nitrodibenzo[b,e][1,4]diazepin-11-yl)benzoic acid.
79. The method and/or the transplantable hematopoietic cell preparation of
any of claims 74, 75 and 76, wherein said Vitamin D receptor antagonist is
selected
from the group consisting of: 1 alpha, 25-(OH)-D3-26,23 lactone; lalpha, 25-
dihydroxyvitamin D (3); the 25-carboxylic ester ZK159222; (23S)- 25-dehydro-1
alpha-OH-D (3); (23R)-25-dehydro-1 alpha-OH-D (3); 1 beta, 25 (OH)2 D3; 1
beta,
25(OH)2-3-epi-D3; (23S) 25-dehydro-1 alpha(OH) D3-26,23-lactone; (23R) 25-
dehydro-1 alpha(OH)D3-26,23-lactone and Butyl-(5Z,7E,22E-(1S,7E,22E-
(1S,3R,24R)-1,3,24-trihydroxy-26,27-cyclo-9,10-secocholesta-5,7,10(19),22-
tetraene-
25-carboxylate).
80. A method of ex-vivo expanding a population of hematopoietic stem
cells, while at the same time, substantially inhibiting differentiation of the
hematopoietic stem cells ex-vivo, the method comprising providing
hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells, with ex-vivo
culture
conditions for ex-vivo cell proliferation and with nicotinamide, a
nicotinamide analog,
a nicotinamide or a nicotinamide analog derivative or a nicotinamide or a
nicotinamide analog metabolite thereby expanding a population of said
hematopoietic
stem cells while at the same time, substantially inhibiting differentiation of
said
hematopoietic stem cells ex-vivo.
81. A method of hematopoietic cells transplantation or implantation
comprising:
(a) obtaining hematopoietic mononuclear cells which comprise a major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells from a donor;
(b) providing said hematopoietic mononuclear cells with ex-vivo culture
conditions for cell proliferation and with nicotinamide, a nicotinamide
analog, a
nicotinamide or a nicotinamide analog derivative or a nicotinamide or a
nicotinamide




116


analog metabolite, thereby expanding a population of said hematopoietic stem
cells,
while at the same time, substantially inhibiting differentiation of said
hematopoietic
stem cells ex-vivo; and
(c) transplanting or implanting said hematopoietic stem cells to a recipient.
82. The method of claim 81, wherein said donor and said recipient are a
single individual.
83. A method of genetically modifying hematopoietic stem cells with an
exogene comprising:
(a) obtaining hematopoietic mononuclear cells which comprise a major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells;
(b) providing said hematopoietic mononuclear cells with ex-vivo culture
conditions for cell proliferation and with nicotinamide, a nicotinamide
analog, a
nicotinamide or a nicotinamide analog derivative or a nicotinamide or a
nicotinamide
analog metabolite, thereby expanding a population of said hematopoietic stem
cells,
while at the same time, substantially inhibiting differentiation of said
hematopoietic
stem cells ex-vivo; and
(c) genetically modifying said hematopoietic stem cells with the exogene.
84. The method of claim 83, wherein genetically modifying is effected by
a vector which comprises the exogene.
85. The method of claim 84, wherein the vector is a viral vector or a
nucleic acid vector.
86. A method of adoptive immunotherapy comprising:
(a) obtaining hematopoietic mononuclear cells which comprise a major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells from a recipient;
(b) providing said hematopoietic mononuclear cells with ex-vivo culture
conditions for cell proliferation and with nicotinamide, a nicotinamide
analog, a




117


nicotinamide or a nicotinamide analog derivative or a nicotinamide or a
nicotinamide
analog metabolite, thereby expanding a population of said hematopoietic stem
cells,
while at the same time, substantially inhibiting differentiation of said
hematopoietic
stem cells; and
(c) transplanting said hematopoietic stem cells to the recipient.
87. A transplantable hematopoietic cell preparation comprising an
expanded population of hematopoietic stem cells propagated ex-vivo from
hematopoietic mononuclear cells which comprise, prior to expansion, a major
fraction
of hematopoietic committed cells and a minor fraction of hematopoietic stem
and
progenitor cells in the presence of an effective amount of an agent selected
from the
group consisting of nicotinamide, a nicotinamide analog, a nicotinamide or a
nicotinamide analog derivative or a nicotinamide or a nicotinamide analog
metabolite,
while at the same time, substantially inhibiting differentiation of said
hematopoietic
stem cells, and a pharmaceutically acceptable carrier.
88. The method and/or the transplantable hematopoietic cell preparation of
any of claims 80, 81, 83, 86 and 87, wherein said hematopoietic mononuclear
cells are
derived from a source selected from the group consisting of bone marrow,
peripheral
blood and neonatal umbilical cord blood.
89. The method of any of claims 80, 81, and 86, wherein providing said
hematopoietic mononuclear cells with said conditions for ex-vivo cell
proliferation
comprises providing said hematopoietic mononuclear cells with nutrients and
with
cytokines.
90. The method of claim 89, wherein said cytokines are early acting
cytokines.
91. The method of claim 90, wherein said early acting cytokines are
selected from the group consisting of stem cell factor, FLT3 ligand,
interleukin-l,
interleukin-2, interleukin-3, interleukin-6, interleukin-10, interleukin-12,
tumor
necrosis factor-a and thrombopoietin.




118


92. The method of claim 89, wherein said cytokines are late acting
cytokines.
93. The method of claim 92, wherein said late acting cytokines are
selected from the group consisting of granulocyte colony stimulating factor,
granulocyte/macrophage colony stimulating factor, erythropoietin, FGF, EGF,
NGF,
VEGF, LIF, Hepatocyte growth factor and macrophage colony stimulating factor.
94. The method and/or the transplantable hematopoietic cell preparation of
any of claims 80, 81, 83, 86 and 87, wherein said nicotinamide analog is
selected
from the group consisting of benzamide, nicotinethioamide, nicotinic acid and
oc-
amino-3-indolepropionic acid.
95. A method of ex-vivo expanding a population of hematopoietic stem
cells, while at the same time, substantially inhibiting differentiation of the
hematopoietic stem cells ex-vivo, the method comprising providing
hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells, with ex-vivo
culture
conditions for ex-vivo cell proliferation and, at the same time, for reducing
an
expression and/or activity of PI 3-kinase, thereby expanding a population of
said
hematopoietic stem cells while at the same time, substantially inhibiting
differentiation of said hematopoietic stem cells ex-vivo.
96. A method of hematopoietic cells transplantation or implantation
comprising:
(a) obtaining hematopoietic mononuclear cells which comprise a major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells from a donor;
(b) providing said hematopoietic mononuclear cells with ex-vivo culture
conditions for cell proliferation and, at the same time, for reducing an
expression
and/or activity of PI 3-kinase, thereby expanding a population of said
hematopoietic
stem cells, while at the same time, substantially inhibiting differentiation
of said
hematopoietic stem cells ex-vivo; and



119


(c) transplanting or implanting said hematopoietic stem cells to a recipient.
97. The method of claim 96, wherein said donor and said recipient are a
single individual.
98. A method of genetically modifying hematopoietic stem cells with an
exogene comprising:
(a) obtaining hematopoietic mononuclear cells which comprise a major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells;
(b) providing said hematopoietic mononuclear cells with ex-vivo culture
conditions for cell proliferation and, at the same time, for reducing an
expression
and/or activity of PI 3-kinase, thereby expanding a population of said
hematopoietic
stem cells, while at the same time, substantially inhibiting differentiation
of said
hematopoietic stem cells ex-vivo; and
(c) genetically modifying said hematopoietic stem cells with the exogene.
99. The method of claim 98, wherein genetically modifying is effected by
a vector which comprises the exogene.
100. The method of claim 99, wherein the vector is a viral vector or a
nucleic acid vector.
101. A method of adoptive immunotherapy comprising:
(a) obtaining hematopoietic mononuclear cells which comprise a major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells from a recipient;
(b) providing said hematopoietic mononuclear cells with ex-vivo culture
conditions for cell proliferation and, at the same time, for reducing an
expression
and/or activity of PI 3-kinase, thereby expanding a population of said
hematopoietic
stem cells, while at the same time, substantially inhibiting differentiation
of said
hematopoietic stem cells; and
(c) transplanting said hematopoietic stem cells to the recipient.




120


102. A transplantable hematopoietic cell preparation comprising an
expanded population of hematopoietic stem cells propagated ex-vivo from
hematopoietic mononuclear cells which comprise, prior to expansion, a major
fraction
of hematopoietic committed cells and a minor fraction of hematopoietic stem
and
progenitor cells in the presence of an effective amount of an agent, said
agent
reducing an expression and/or activity of PI 3-kinase, while at the same time,
substantially inhibiting differentiation of said hematopoietic stem cells, and
a
pharmaceutically acceptable carrier.
103. The method of any of claims 95, 96, 98, and 101, wherein said
hematopoietic mononuclear cells are derived from a source selected from the
group
consisting of bone marrow, peripheral blood and neonatal umbilical cord blood.
104. The method of any of claims 95, 96, 98 and 101, wherein providing
said hematopoietic mononuclear cells with said conditions for ex-vivo cell
proliferation comprises providing said hematopoietic mononuclear cells with
nutrients
and with cytokines.
105. The method of claim 104, wherein said cytokines are early acting
cytokines.
106. The method of claim 105, wherein said early acting cytokines are
selected from the group consisting of stem cell factor, FLT3 ligand,
interleukin-1,
interleukin-2, interleukin-3, interleukin-6, interleukin-10, interleukin-12,
tumor
necrosis factor-a and thrombopoietin.
107. The method of claim 104, wherein said cytokines are late acting
cytokines.
108. The method of claim 107, wherein said late acting cytokines are
selected from the group consisting of granulocyte colony stimulating factor,
granulocyte/macrophage colony stimulating factor, erythropoietin, FGF, EGF,
NGF,
VEGF, LIF, Hepatocyte growth factor and macrophage colony stimulating factor.




121


109. The method of any of claims 95, 96, 98 and 101, wherein providing
said hematopoietic mononuclear cells with ex-vivo culture conditions for
reducing
said expression and/or said activity of PI 3-kinase is by providing said
hematopoietic
mononuclear cells with an agent that downregulates PI 3-kinase expression.
110. The transplantable hematopoietic cell preparation of claim 102,
wherein said agent is an agent that downregulates PI 3-kinase expression.
111. The method and/or transplantable hematopoietic cell preparation of
any claim 109 and 110, wherein said agent that downregulates PI 3-kinase
expression is a polynucleotide.
112. The method and/or transplantable hematopoietic cell preparation of
any claim 109 and 110, wherein said agent that downregulates PI 3-kinase
expression is an intracellular antibody.
113. The method of claim 112, wherein said polynucleotide is a small
interfering polynucleotide molecule directed to cause intracellular PI 3-
kinase
mRNA or gene degradation.
114. The method of claim 113, wherein said small interfering
polynucleotide molecule is selected from the group consisting of an RNAi
molecule,
an anti-sense molecule, a rybozyme molecule and a DNAzyme molecule.
115. The method and/or transplantable hematopoietic cell preparation of
any claim 109 and 110, wherein providing said hematopoietic mononuclear cells
with ex-vivo culture conditions for reducing said expression and/or said
activity of PI
3-kinase is by providing said hematopoietic mononuclear cells with an agent
that
inhibits PI 3-kinase activity.
116. The transplantable hematopoietic cell preparation of claim 102,
wherein said agent is an agent that inhibits PI 3-kinase activity.




122


117. The method and/or the transplantable hematopoietic cell preparation
of any of claims 115 and 116, wherein said agent that inhibits PI 3-kinase
activity is
selected from the group consisting of wortmannin and LY294002
118. A method of ex-vivo expanding a population of hematopoietic stem
cells, while at the same time, substantially inhibiting differentiation of the
hematopoietic stem cells ex-vivo, the method comprising providing
hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells, with ex-vivo
culture
conditions for ex-vivo cell proliferation and, at the same time, with at least
one copper
chelator or chelate, thereby expanding a population of said hematopoietic stem
cells
while at the same time, substantially inhibiting differentiation of said
hematopoietic
stem cells ex-vivo.
119. A method of hematopoietic cells transplantation or implantation
comprising:
(a) obtaining hematopoietic mononuclear cells which comprise a major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells from a donor;
(b) providing said hematopoietic mononuclear cells with ex-vivo culture
conditions for cell proliferation and, at the same time, with at least one
copper
chelator or chelate, thereby expanding a population of said hematopoietic stem
cells,
while at the same time, substantially inhibiting differentiation of said
hematopoietic
stem cells ex-vivo; and
(c) transplanting or implanting said hematopoietic stem cells to a recipient.
120. The method of claim 119, wherein said donor and said recipient are a
single individual.
121. A method of genetically modifying hematopoietic stem cells with an
exogene comprising:




123


(a) obtaining hematopoietic mononuclear cells which comprise a major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells;
(b) providing said hematopoietic mononuclear cells with ex-vivo culture
conditions for cell proliferation and, at the same time, with at least one
copper
chelator or chelate, thereby expanding a population of said hematopoietic stem
cells,
while at the same time, substantially inhibiting differentiation of said
hematopoietic
stem cells ex-vivo; and
(c) genetically modifying said hematopoietic stem cells with the exogene.
122. The method of claim 121, wherein genetically modifying is effected by
a vector which comprises the exogene.
123. The method of claim 122, wherein the vector is a viral vector or a
nucleic acid vector.
124. A method of adoptive immunotherapy comprising:
(a) obtaining hematopoietic mononuclear cells which comprise a major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells from a recipient;
(b) providing said hematopoietic mononuclear cells with ex-vivo culture
conditions for cell proliferation and, at the same time, with at least one
copper
chelator or chelate, thereby expanding a population of said hematopoietic stem
cells,
while at the same time, substantially inhibiting differentiation of said
hematopoietic
stem cells; and
(c) transplanting said hematopoietic stem cells to the recipient.
125. A transplantable hematopoietic cell preparation comprising an
expanded population of hematopoietic stem cells propagated ex-vivo from
hematopoietic mononuclear cells which comprise, prior to expansion, a major
fraction
of hematopoietic committed cells and a minor fraction of hematopoietic stem
and
progenitor cells in the presence of an effective amount of at least one copper
chelate




124


or chelator, while at the same time, substantially inhibiting differentiation
of said
hematopoietic stem cells, and a pharmaceutically acceptable carrier.
126. The method and/or the transplantable hematopoietic cell preparation of
any of claims 118, 119, 121, 124 and 125, wherein said hematopoietic
mononuclear
cells are derived from a source selected from the group consisting of bone
marrow,
peripheral blood and neonatal umbilical cord blood.
127. The method of any of claims 118, 119, and 124, wherein providing
said hematopoietic mononuclear cells with said conditions for ex-vivo cell
proliferation comprises providing said hematopoietic mononuclear cells with
nutrients
and with cytokines.
128. The method of claim 127, wherein said cytokines are early acting
cytokines.
129. The method of claim 128, wherein said early acting cytokines are
selected from the group consisting of stem cell factor, FLT3 ligand,
interleukin-1,
interleukin-2, interleukin-3, interleukin-6, interleukin-10, interleukin-12,
tumor
necrosis factor-a and thrombopoietin.
130. The method of claim 127, wherein said cytokines are late acting
cytokines.
131. The method of claim 130, wherein said late acting cytokines are
selected from the group consisting of granulocyte colony stimulating factor,
granulocyte/macrophage colony stimulating factor, erythropoietin, FGF, EGF,
NGF,
VEGF, LIF, Hepatocyte growth factor and macrophage colony stimulating factor.
132. The method of any of claims 118, 119, 121 and 124, wherein providing
said hematopoietic mononuclear cells with at least one copper chelator or
chelate is
by providing said hematopoietic mononuclear cells at least one copper
chelator.




125

133. The method of claim 132, further comprising providing said
hematopoietic mononuclear cells fraction zinc.
134. The transplantable hematopoietic cell preparation of claim 125,
wherein said expanded population of said hematopoietic stem cells fraction is
propagated ex-vivo from hematopoietic mononuclear cells which comprise a major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells in the presence of an effective amount of at least one
copper
chelator.
135. The transplantable hematopoietic cell preparation of claim 134,
wherein said expanded population of said hematopoietic stem cells fraction is
propagated ex-vivo from hematopoietic mononuclear cells which comprise a major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells in the presence of an effective amount of zinc.
136. An assay of determining whether a transition metal chelate or chelator
causes substantial inhibition or induction of differentiation of hematopoietic
stem
cells, the assay comprising:
culturing hematopoietic mononuclear cells which comprise a major fraction of
hematopoietic committed cells and a minor fraction of hematopoietic stem and
progenitor cells, in the presence of the transition metal chelate or chelator
and
monitoring differentiation of said hematopoietic stem cells, wherein if
differentiation
is increased as is compared to non-treated hematopoietic mononuclear cells,
said
transition metal chelate induces differentiation, whereas if differentiation
is decreased
as is compared to non-treated hematopoietic mononuclear cells, or if
differentiation is
absent altogether, said transition metal chelate inhibits differentiation.
137. The method, the assay and/or the transplantable hematopoietic cell
preparation of any of claims 118, 119, 121, 124, 125, 136, and 138-141,
wherein said
at least one copper chelate or chelator comprises a polyamine chelator.





126

138. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 137, wherein said polyamine chelator is capable of
forming an
organometallic complex with a transition metal other than copper.
139. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 138, wherein said transition metal is selected from the
group
consisting of zinc, cobalt, nickel, iron, palladium, platinum, rhodium and
ruthenium.
140. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 137, wherein said polyamine chelator is a linear
polyamine.
141. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 140, wherein said linear polyamine has a general formula
I:

HX-Am-(Y1B1)1.....(YnBn)n-ZH

Formula I
wherein:
m is an integer from 1 to 10;
n is an integer from 0 to 20;
X and Z are each independently selected from the group consisting of an
oxygen atom, a sulfur atom and a -NH group;
Y1 and Yn are each independently selected from the group consisting of an
oxygen atom, a sulfur atom and a -NH group;
A is an alkylene chain having between 1 and 10 substituted and/or non-
substituted carbon atoms; and
B, and Bn are each independently an alkylene chain having between 1 and 20
substituted and/or non-substituted carbon atoms,
provided that at least one of said X, Z, Y1 and Yn is a -NH group and/or at
least one of said carbon atoms in said alkylene chains is substituted by an
amine
group.




127

142. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 141, wherein said A is an alkylene chain having a general
formula II:
Image
wherein:
g is an integer that equals 0 or 3-10;
each of R1, R2 and Rg is independently selected from the group consisting of
hydrogen, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroalicyclic,
heteroaryl, halo,
amino, alkylamino, arylamino, cycloalkylamino, heteroalicyclic amino,
heteroarylamino, hydroxy, alkoxy, aryloxy, azo, C-amido, N-amido, ammonium,
thiohydroxy, thioalkoxy, thioaryloxy, sulfonyl, sulfinyl, N-sulfonamide, S-
sulfonamide, phosphonyl, phosphinyl, phosphonium, carbonyl, thiocarbonyl, C-
carboxy, O-carboxy, C-thiocarboxy, O-thiocarboxy, N-carbamate, O-carbamate, N-
thiocarbamate, O-thiocarbamate, urea, thiourea, borate, borane, boroaza,
silyl, siloxy,
silaza, aquo, alcohol, peroxo, amine oxide, hydrazine, alkyl hydrazine, aryl
hydrazine,
nitric oxide, cyanate, thiocyanate, isocyanate, isothiocyanate, cyano,
alkylnitrile, aryl
nitrile, alkyl isonitrile, aryl isonitrile, nitrate, nitrite, azido, alkyl
sulfonic acid, aryl
sulfonic acid, alkyl sulfoxide, aryl sulfoxide, alkyl aryl sulfoxide, alkyl
sulfenic acid,
aryl sulfenic acid, alkyl sulfinic acid, aryl sulfuric acid, alkyl thiol
carboxylic acid,
aryl thiol carboxylic acid, alkyl thiol thiocarboxylic acid, aryl thiol
thiocarboxylic
acid, carboxylic acid, alkyl carboxylic acid, aryl carboxylic acid, sulfate,
sulfite,
bisulfate, thiosulfate, thiosulfite, alkyl phosphine, aryl phosphine, alkyl
phosphine
oxide, aryl phosphine oxide, alkyl aryl phosphine oxide, alkyl phosphine
sulfide, aryl
phosphine sulfide, alkyl aryl phosphine sulfide, alkyl phosphonic acid, aryl
phosphonic acid, alkyl phosphinic acid, aryl phosphinic acid, phosphate,
thiophosphate, phosphate, pyrophosphate, triphosphate, hydrogen phosphate,
dihydrogen phosphate, guanidino, S-dithiocarbamate, N-dithiocarbamate,
bicarbonate,
carbonate, perchlorate, chlorate, chlorite, hypochlorite, perbromate, bromate,
bromite,
hypobromite, tetrahalomanganate, tetrafluoroborate, hexafluoroantimonate,





128

hypophosphite, iodate, periodate, metaborate, tetraarylborate, tetraalkyl
borate,
tartarate, salicylate, succinate, citrate, ascorbate, saccharirate, amino
acid, hydroxamic
acid and thiotosylate.

143. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 142, wherein each of B1 and Bn is independently an
alkylene
chain having a general formula III:
Image
wherein:
p is an integer that equals 0 or g+1;
q is an integer from g+2 to g+20; and
each of Rp, Rp+1 and Rq is independently selected from the group consisting
of hydrogen, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroalicyclic,
heteroaryl, halo,
amino, alkylamino, arylamino, cycloalkylamino, heteroalicyclic amino,
heteroarylamino, hydroxy, alkoxy, aryloxy, azo, C-amido, N-amido, ammonium,
thiohydroxy, thioalkoxy, thioaryloxy, sulfonyl, sulfinyl, N-sulfonamide, S-
sulfonamide, phosphonyl, phosphinyl, phosphonium, carbonyl, thiocarbonyl, C-
carboxy, O-carboxy, C-thiocarboxy, O-thiocarboxy, N-carbamate, O-carbamate, N-
thiocarbamate, O-thiocarbamate, urea, thiourea, borate, borane, boroaza,
silyl, siloxy,
silaza, aquo, alcohol, peroxo, amine oxide, hydrazine, alkyl hydrazine, aryl
hydrazine,
nitric oxide, cyanate, thiocyanate, isocyanate, isothiocyanate, cyano,
alkylnitrile, aryl
nitrite, alkyl isonitrile, aryl isonitrile, nitrate, nitrite, azido, alkyl
sulfonic acid, aryl
sulfonic acid, alkyl sulfoxide, aryl sulfoxide, alkyl aryl sulfoxide, alkyl
sulfenic acid,
aryl sulfenic acid, alkyl sulfinic acid, aryl sulfinic acid, alkyl thiol
carboxylic acid,
aryl thiol carboxylic acid, alkyl thiol thiocarboxylic acid, aryl thiol
thiocarboxylic
acid, carboxylic acid, alkyl carboxylic acid, aryl carboxylic acid, sulfate,
sulfite,
bisulfate, thiosulfate, thiosulfite, alkyl phosphine, aryl phosphine, alkyl
phosphine
oxide, aryl phosphine oxide, alkyl aryl phosphine oxide, alkyl phosphine
sulfide, aryl
phosphine sulfide, alkyl aryl phosphine sulfide, alkyl phosphonic acid, aryl
phosphonic acid, alkyl phosphinic acid, aryl phosphinic acid, phosphate,





129

thiophosphate, phosphite, pyrophosphite, triphosphate, hydrogen phosphate,
dihydrogen phosphate, guanidino, S-dithiocarbamate, N-dithiocarbamate,
bicarbonate,
carbonate, perchlorate, chlorate, chlorite, hypochlorite, perbromate, bromate,
bromite,
hypobromite, tetrahalomanganate, tetrafluoroborate, hexafluoroantimonate,
hypophosphite, iodate, periodate, metaborate, tetraarylborate, tetraalkyl
borate,
tartarate, salicylate, succinate, citrate, ascorbate, saccharirate, amino
acid, hydroxamic
acid and thiotosylate.

144. The method, the assay, the pharmaceutical composition, the kit, the
expanded population and/or the assay of claim 141, wherein said linear
polyamine is
tetraethylenepentamine.

145. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 142, wherein at least one of said C1, C2 and Cg is a
chiral carbon
atom.

146. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 143, wherein at least one of said Cp, Cp+1 and Cq is a
chiral
carbon atom.

147. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 137, wherein said polyamine chelator is a cyclic
polyamine.

148. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 147, wherein said cyclic polyamine is cyclam.

149. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 147, wherein said cyclic polyamine has a general formula
IV:

Image




130

wherein:
m is an integer from 1 to 10;
n is an integer from 0 to 20;
X and Z are each independently selected from the group consisting of an
oxygen atom, a sulfur atom and a -NH group;
Y1 and Yn are each independently selected from the group consisting of an
oxygen atom, a sulfur atom and a -NH group;
A is an alkylene chain having between 1 and 10 substituted and/or non-
substituted carbon atoms;
B1 and Bn are each independently an alkylene chain having between 1 and 20
substituted and/or non-substituted carbon atoms; and
D is a bridging group having a general formula V:

U-W-V
Formula V

whereas:
U and V are each independently selected from the group consisting of
substituted hydrocarbon chain and non-substituted hydrocarbon chain; and
W is selected from the group consisting of amide, ether, ester, disulfide,
thioether, thioester, imine and alkene,
provided that at least one of said X, Z, Y1 and Yn is a -NH group and/or at
least one of said carbon atoms in said alkylene chains is substituted by an
amine
group.

150. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 149, wherein said A is an alkylene chain having a general
formula II:
Image




131

wherein:
g is an integer that equals 0 or 3-10; and
each of R1, R2 and Rg is independently selected from the group consisting of
hydrogen, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroalicyclic,
heteroaryl, halo,
amino, alkylamino, arylamino, cycloalkylamino, heteroalicyclic amino,
heteroarylamino, hydroxy, alkoxy, aryloxy, azo, C-amido, N-amido, ammonium,
thiohydroxy, thioalkoxy, thioaryloxy, sulfonyl, sulfinyl, N-sulfonamide, S-
sulfonamide, phosphonyl, phosphinyl, phosphonium, carbonyl, thiocarbonyl, C-
carboxy, O-carboxy, C-thiocarboxy, O-thiocarboxy, N-carbamate, O-carbamate, N-
thiocarbamate, O-thiocarbamate, urea, thiourea, borate, borane, boroaza,
silyl, siloxy,
silaza, aquo, alcohol, peroxo, amine oxide, hydrazine, alkyl hydrazine, aryl
hydrazine,
nitric oxide, cyanate, thiocyanate, isocyanate, isothiocyanate, cyano,
alkylnitrile, aryl
nitrile, alkyl isonitrile, aryl isonitrile, nitrate, nitrite, azido, alkyl
sulfonic acid, aryl
sulfonic acid, alkyl sulfoxide, aryl sulfoxide, alkyl aryl sulfoxide, alkyl
sulfenic acid,
aryl sulfenic acid, alkyl sulfinic acid, aryl sulfinic acid, alkyl thiol
carboxylic acid,
aryl thiol carboxylic acid, alkyl thiol thiocarboxylic acid, aryl thiol
thiocarboxylic
acid, carboxylic acid, alkyl carboxylic acid, aryl carboxylic acid, sulfate,
sulfite,
bisulfate, thiosulfate, thiosulfite, alkyl phosphine, aryl phosphine, alkyl
phosphine
oxide, aryl phosphine oxide, alkyl aryl phosphine oxide, alkyl phosphine
sulfide, aryl
phosphine sulfide, alkyl aryl phosphine sulfide, alkyl phosphonic acid, aryl
phosphonic acid, alkyl phosphinic acid, aryl phosphinic acid, phosphate,
thiophosphate, phosphate, pyrophosphate, triphosphate, hydrogen phosphate,
dihydrogen phosphate, guanidino, S-dithiocarbamate, N-dithiocarbamate,
bicarbonate,
carbonate, perchlorate, chlorate, chlorite, hypochlorite, perbromate, bromate,
bromite,
hypobromite, tetrahalomanganate, tetrafluoroborate, hexafluoroantimonate,
hypophosphite, iodate, periodate, metaborate, tetraarylborate, tetraalkyl
borate,
tartarate, salicylate, succinate, citrate, ascorbate, saccharirate, amino
acid, hydroxamic
acid and thiotosylate.

151. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 150, wherein each of B1 and Bn is independently an
alkylene
chain having a general formula III:




Image

wherein:
p is an integer that equals 0 or g+1;
q is an integer from g+2 to g+20; and
each of Rp, Rp+1 and Rq is independently selected from the group consisting
of hydrogen, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroalicyclic,
heteroaryl, halo,
amino, alkylamino, arylamino, cycloalkylamino, heteroalicyclic amino,
heteroarylamino, hydroxy, alkoxy, aryloxy, azo, C-amido, N-amido, ammonium,
thiohydroxy, thioalkoxy, thioaryloxy, sulfonyl, sulfinyl, N-sulfonamide, S-
sulfonamide, phosphonyl, phosphinyl, phosphonium, carbonyl, thiocarbonyl, C-
carboxy, O-carboxy, C-thiocarboxy, O-thiocarboxy, N-carbamate, O-carbamate, N-
thiocarbamate, O-thiocarbamate, urea, thiourea, borate, borane, boroaza,
silyl, siloxy,
silaza, aquo, alcohol, peroxo, amine oxide, hydrazine, alkyl hydrazine, aryl
hydrazine,
nitric oxide, cyanate, thiocyanate, isocyanate, isothiocyanate, cyano,
alkylnitrile, aryl
nitrite, alkyl isonitrile, aryl isonitrile, nitrate, nitrite, azido, alkyl
sulfonic acid, aryl
sulfonic acid, alkyl sulfoxide, aryl sulfoxide, alkyl aryl sulfoxide, alkyl
sulfenic acid,
aryl sulfenic acid, alkyl sulfinic acid, aryl sulfuric acid, alkyl thiol
carboxylic acid,
aryl thiol carboxylic acid, alkyl thiol thiocarboxylic acid, aryl thiol
thiocarboxylic
acid, carboxylic acid, alkyl carboxylic acid, aryl carboxylic acid, sulfate,
sulfite,
bisulfate, thiosulfate, thiosulfite, alkyl phosphine, aryl phosphine, alkyl
phosphine
oxide, aryl phosphine oxide, alkyl aryl phosphine oxide, alkyl phosphine
sulfide, aryl
phosphine sulfide, alkyl aryl phosphine sulfide, alkyl phosphonic acid, aryl
phosphonic acid, alkyl phosphinic acid, aryl phosphinic acid, phosphate,
thiophosphate, phosphate, pyrophosphate, triphosphate, hydrogen phosphate,
dihydrogen phosphate, guanidino, S-dithiocarbamate, N-dithiocarbamate,
bicarbonate,
carbonate, perchlorate, chlorate, chlorite, hypochlorite, perbromate, bromate,
bromite,
hypobromite, tetrahalomanganate, tetrafluoroborate, hexafluoroantimonate,
hypophosphite, iodate, periodate, metaborate, tetraarylborate, tetraalkyl
borate,




133

tartarate, salicylate, succinate, citrate, ascorbate, saccharirate, amino
acid, hydroxamic
acid and thiotosylate.

152. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 150, wherein at least one of said C1, C2 and Cg is a
chiral carbon
atom.

153. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 151, wherein at least one of said Cp, Cp+1 and Cq is a
chiral
carbon atom.

154. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 147, wherein said cyclic polyamine has a general formula
selected from the group consisting of:

Image




Image
wherein:
m is an integer from 1 to 10;
n is an integer from 0 to 20;
X and Z are each independently selected from the group consisting of an
oxygen atom, a sulfur atom and a -NH group;
Y1 and Yn are each independently selected from the group consisting of an
oxygen atom, a sulfur atom and a -NH group;
A is an alkylene chain having between 1 and 10 substituted and/or non-
substituted carbon atoms;
B1 and Bn are each independently an alkylene chain having between 1 and 20
substituted and/or non-substituted carbon atoms; and
D is a bridging group having a general formula V:

U-W-V
Formula V

whereas:
U and V are each independently selected from the group consisting of
substituted hydrocarbon chain and non-substituted hydrocarbon chain; and
W is selected from the group consisting of amide, ether, ester, disulfide,
thioether, thioester, imine and alkene,
and further wherein should said D is attached at one end to A (Formulas VI,
VII and X), said U or said V are being attached to one carbon atom in said
alkylene
chain and should said D is attached at one end to B1 or Bn (Formulas VIII, IX
and X),
said U or said V are being attached to one carbon atom in said alkylene chain,
provided that at least one of said X, Z, Y1 and Yn is a -NH group and/or at
least one of said carbon atoms in said alkylene chains is substituted by an
amine
group.




135
155. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 154, wherein said A is an alkylene chain having a general
formula II:
Image
wherein:
g is an integer that equals 0 or 3-10; and
each of R1, R2 and Rg is independently selected from the group consisting of
hydrogen, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroalicyclic,
heteroaryl, halo,
amino, alkylamino, arylamino, cycloalkylamino, heteroalicyclic amino,
heteroarylamino, hydroxy, alkoxy, aryloxy, azo, C-amido, N-amido, ammonium,
thiohydroxy, thioalkoxy, thioaryloxy, sulfonyl, sulfinyl, N-sulfonamide, S-
sulfonamide, phosphonyl, phosphinyl, phosphonium, carbonyl, thiocarbonyl, C-
carboxy, O-carboxy, C-thiocarboxy, O-thiocarboxy, N-carbamate, O-carbamate, N-
thiocarbamate, O-thiocarbamate, urea, thiourea, borate, borane, boroaza,
silyl, siloxy,
silaza, aquo, alcohol, peroxo, amine oxide, hydrazine, alkyl hydrazine, aryl
hydrazine,
nitric oxide, cyanate, thiocyanate, isocyanate, isothiocyanate, cyano,
alkylnitrile, aryl
nitrile, alkyl isonitrile, aryl isonitrile, nitrate, nitrite, azido, alkyl
sulfonic acid, aryl
sulfonic acid, alkyl sulfoxide, aryl sulfoxide, alkyl aryl sulfoxide, alkyl
sulfenic acid,
aryl sulfenic acid, alkyl sulfinic acid, aryl sulfinic acid, alkyl thiol
carboxylic acid,
aryl thiol carboxylic acid, alkyl thiol thiocarboxylic acid, aryl thiol
thiocarboxylic
acid, carboxylic acid, alkyl carboxylic acid, aryl carboxylic acid, sulfate,
sulfite,
bisulfate, thiosulfate, thiosulfite, alkyl phosphine, aryl phosphine, alkyl
phosphine
oxide, aryl phosphine oxide, alkyl aryl phosphine oxide, alkyl phosphine
sulfide, aryl
phosphine sulfide, alkyl aryl phosphine sulfide, alkyl phosphonic acid, aryl
phosphonic acid, alkyl phosphinic acid, aryl phosphinic acid, phosphate,
thiophosphate, phosphate, pyrophosphate, triphosphate, hydrogen phosphate,
dihydrogen phosphate, guanidino, S-dithiocarbamate, N-dithiocarbamate,
bicarbonate,
carbonate, perchlorate, chlorate, chlorite, hypochlorite, perbromate, bromate,
bromite,
hypobromite, tetrahalomanganate, tetrafluoroborate, hexafluoroantimonate,
hypophosphite, iodate, periodate, metaborate, tetraarylborate, tetraalkyl
borate,




136

tartarate, salicylate, succinate, citrate, ascorbate, saccharirate, amino
acid, hydroxamic
acid and thiotosylate.

156. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 155, wherein each of B1 and Bn is independently an
alkylene
chain having a general formula III:
Image
wherein:
p is an integer that equals 0 or g+1;
q is an integer from g+2 to g+20; and
each of Rp, Rp+1 and Rq is independently selected from the group consisting
of hydrogen, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroalicyclic,
heteroaryl, halo,
amino, alkylamino, arylamino, cycloalkylamino, heteroalicyclic amino,
heteroarylamino, hydroxy, alkoxy, aryloxy, azo, C-amido, N-amido, ammonium,
thiohydroxy, thioalkoxy, thioaryloxy, sulfonyl, sulfinyl, N-sulfonamide, S-
sulfonamide, phosphonyl, phosphinyl, phosphonium, carbonyl, thiocarbonyl, C-
carboxy, O-carboxy, C-thiocarboxy, O-thiocarboxy, N-carbamate, O-carbamate, N-
thiocarbamate, O-thiocarbamate, urea, thiourea, borate, borane, boroaza,
silyl, siloxy,
silaza, aquo, alcohol, peroxo, amine oxide, hydrazine, alkyl hydrazine, aryl
hydrazine,
nitric oxide, cyanate, thiocyanate, isocyanate, isothiocyanate, cyano,
alkylnitrile, aryl
nitrite, alkyl isonitrile, aryl isonitrile, nitrate, nitrite, azido, alkyl
sulfonic acid, aryl
sulfonic acid, alkyl sulfoxide, aryl sulfoxide, alkyl aryl sulfoxide, alkyl
sulfenic acid,
aryl sulfenic acid, alkyl sulfuric acid, aryl sulfinic acid, alkyl thiol
carboxylic acid,
aryl thiol carboxylic acid, alkyl thiol thiocarboxylic acid, aryl thiol
thiocarboxylic
acid, carboxylic acid, alkyl carboxylic acid, aryl carboxylic acid, sulfate,
sulfite,
bisulfate, thiosulfate, thiosulfite, alkyl phosphine, aryl phosphine, alkyl
phosphine
oxide, aryl phosphine oxide, alkyl aryl phosphine oxide, alkyl phosphine
sulfide, aryl
phosphine sulfide, alkyl aryl phosphine sulfide, alkyl phosphonic acid, aryl
phosphonic acid, alkyl phosphinic acid, aryl phosphinic acid, phosphate,
thiophosphate, phosphate, pyrophosphate, triphosphate, hydrogen phosphate,





137

dihydrogen phosphate, guanidino, S-dithiocarbamate, N-dithiocarbamate,
bicarbonate,
carbonate, perchlorate, chlorate, chlorite, hypochlorite, perbromate, bromate,
bromite,
hypobromite, tetrahalomanganate, tetrafluoroborate, hexafluoroantimonate,
hypophosphite, iodate, periodate, metaborate, tetraarylborate, tetraalkyl
borate,
tartarate, salicylate, succinate, citrate, ascorbate, saccharirate, amino
acid, hydroxamic
acid and thiotosylate.

157. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 155, wherein at least one of said C1, C2 and Cg is a
chiral carbon
atom.

158. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 156, wherein at least one of said Cp, Cp+1 and Cq is a
chiral
carbon atom.

159. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 137, wherein said polyamine chelator includes at least
one linear
polyamine and at least one cyclic polyamine.

160. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 159, wherein said polyamine chelator has a general
formula XI:
{E~f ~QlyGl~g~~ti ~~E2~~ ~Q2yCT2~j~}k_......._f~En~l'fQn_(Gn)ol~t
Formula XI
wherein:
n is an integer greater than 1;
each of f, g, h, i, j, k, l, o and t is independently an integer from 0 to 10;
each of E,, EZ and En is independently a linear polyamine;
each of G,, GZ and Gn is independently a cyclic polyamine; and
each of Q,, QZ and Qn is independently a linker linking between two of said
polyamines,




138

provided that at least one of said Q1, Q2 and Qn is an amine group and/or at
least one of said linear polyamine and said cyclic polyamine is having at
least one free
amine group.

161. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 160, wherein each of said Q1, Q2 and Qn is independently
selected from the group consisting alkylene, alkenylene, alkynylene, arylene,
cycloalkylene, hetroarylene, amine, azo, amide, sulfonyl, sulfinyl,
sulfonamide,
phosphonyl, phosphinyl, phosphonium, ketoester, carbonyl, thiocarbonyl, ester,
ether,
thioether, carbamate, thiocarbamate, urea, thiourea, borate, borane, boroaza,
silyl,
siloxy and silaza.

162. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 159, wherein each of said E1, E2 and En is independently
a linear
polyamine having a general formula I:

HX-Am-(Y1B1)1....(YnBn)n-ZH

Formula I

wherein:
m is an integer from 1 to 10;
n is an integer from 0 to 20;
X and Z are each independently selected from the group consisting of an
oxygen atom, a sulfur atom and a -NH group;
Y1 and Yn are each independently selected from the group consisting of an
oxygen atom, a sulfur atom and a -NH group;
A is an alkylene chain having between 1 and 10 substituted and/or non-
substituted carbon atoms; and
B1 and Bn are each independently an alkylene chain having between 1 and 20
substituted and/or non-substituted carbon atoms,
provided that at least one of said X, Z, Y1 and Yn is a -NH group and/or at
least one of said carbon atoms in said alkylene chains is substituted by an
amine
group.




139

163. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 162, wherein said A is an alkylene chain having a general
formula II:
Image
wherein:
g is an integer that equals 0 or 3-10;
each of R1, R2 and Rg is independently selected from the group consisting of
hydrogen, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroalicyclic,
heteroaryl, halo,
amino, alkylamino, arylamino, cycloalkylamino, heteroalicyclic amino,
heteroarylamino, hydroxy, alkoxy, aryloxy, azo, C-amido, N-amido, ammonium,
thiohydroxy, thioalkoxy, thioaryloxy, sulfonyl, sulfinyl, N-sulfonamide, S-
sulfonamide, phosphonyl, phosphinyl, phosphonium, carbonyl, thiocarbonyl, C-
carboxy, O-carboxy, C-thiocarboxy, O-thiocarboxy, N-carbamate, O-carbamate, N-
thiocarbamate, O-thiocarbamate, urea, thiourea, borate, borane, boroaza,
silyl, siloxy,
silaza, aquo, alcohol, peroxo, amine oxide, hydrazine, alkyl hydrazine, aryl
hydrazine,
nitric oxide, cyanate, thiocyanate, isocyanate, isothiocyanate, cyano,
alkylnitrile, aryl
nitrite, alkyl isonitrile, aryl isonitrile, nitrate, nitrite, azido, alkyl
sulfonic acid, aryl
sulfonic acid, alkyl sulfoxide, aryl sulfoxide, alkyl aryl sulfoxide, alkyl
sulfenic acid,
aryl sulfenic acid, alkyl sulfinic acid, aryl sulfinic acid, alkyl thiol
carboxylic acid,
aryl thiol carboxylic acid, alkyl thiol thiocarboxylic acid, aryl thiol
thiocarboxylic
acid, carboxylic acid, alkyl carboxylic acid, aryl carboxylic acid, sulfate,
sulfite,
bisulfate, thiosulfate, thiosulfite, alkyl phosphine, aryl phosphine, alkyl
phosphine
oxide, aryl phosphine oxide, alkyl aryl phosphine oxide, alkyl phosphine
sulfide, aryl
phosphine sulfide, alkyl aryl phosphine sulfide, alkyl phosphonic acid, aryl
phosphonic acid, alkyl phosphinic acid, aryl phosphinic acid, phosphate,
thiophosphate, phosphate, pyrophosphate, triphosphate, hydrogen phosphate,
dihydrogen phosphate, guanidino, S-dithiocarbamate, N-dithiocarbamate,
bicarbonate,
carbonate, perchlorate, chlorate, chlorite, hypochlorite, perbromate, bromate,
bromite,
hypobromite, tetrahalomanganate, tetrafluoroborate, hexafluoroantimonate,
hypophosphite, iodate, periodate, metaborate, tetraarylborate, tetraalkyl
borate,




140

tartarate, salicylate, succinate, citrate, ascorbate, saccharirate, amino
acid, hydroxamic
acid and thiotosylate.

164. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 163, wherein each of B1 and Bn is independently an
alkylene
chain having a general formula III:

Image

wherein:
p is an integer that equals 0 or g+1;
q is an integer from g+2 to g+20; and
each of Rp, Rp+1 and Rq is independently selected from the group consisting
of hydrogen, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroalicyclic,
heteroaryl, halo,
amino, alkylamino, arylamino, cycloalkylamino, heteroalicyclic amino,
heteroarylamino, hydroxy, alkoxy, aryloxy, azo, C-amido, N-amido, ammonium,
thiohydroxy, thioalkoxy, thioaryloxy, sulfonyl, sulfinyl, N-sulfonamide, S-
sulfonamide, phosphonyl, phosphinyl, phosphonium, carbonyl, thiocarbonyl, C-
carboxy, O-carboxy, C-thiocarboxy, O-thiocarboxy, N-carbamate, O-carbamate, N-
thiocarbamate, O-thiocarbamate, urea, thiourea, borate, borane, boroaza,
silyl, siloxy,
silaza, aquo, alcohol, peroxo, amine oxide, hydrazine, alkyl hydrazine, aryl
hydrazine,
nitric oxide, cyanate, thiocyanate, isocyanate, isothiocyanate, cyano,
alkylnitrile, aryl
nitrile, alkyl isonitrile, aryl isonitrile, nitrate, nitrite, azido, alkyl
sulfonic acid, aryl
sulfonic acid, alkyl sulfoxide, aryl sulfoxide, alkyl aryl sulfoxide, alkyl
sulfenic acid,
aryl sulfenic acid, alkyl sulfinic acid, aryl sulfinic acid, alkyl thiol
carboxylic acid,
aryl thiol carboxylic acid, alkyl thiol thiocarboxylic acid, aryl thiol
thiocarboxylic
acid, carboxylic acid, alkyl carboxylic acid, aryl carboxylic acid, sulfate,
sulfite,
bisulfate, thiosulfate, thiosulfite, alkyl phosphine, aryl phosphine, alkyl
phosphine
oxide, aryl phosphine oxide, alkyl aryl phosphine oxide, alkyl phosphine
sulfide, aryl
phosphine sulfide, alkyl aryl phosphine sulfide, alkyl phosphonic acid, aryl





141

phosphonic acid, alkyl phosphinic acid, aryl phosphinic acid, phosphate,
thiophosphate, phosphite, pyrophosphite, triphosphate, hydrogen phosphate,
dihydrogen phosphate, guanidino, S-dithiocarbamate, N-dithiocarbamate,
bicarbonate,
carbonate, perchlorate, chlorate, chlorite, hypochlorite, perbromate, bromate,
bromite,
hypobromite, tetrahalomanganate, tetrafluoroborate, hexafluoroantimonate,
hypophosphite, iodate, periodate, metaborate, tetraarylborate, tetraalkyl
borate,
tartarate, salicylate, succinate, citrate, ascorbate, saccharirate, amino
acid, hydroxamic
acid and thiotosylate.

165. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 163, wherein at least one of said C1, C2 and Cg is a
chiral carbon
atom.

166. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 164, wherein at least one of said Cp, Cp+1 and Cq is a
chiral
carbon atom.

167. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 160, wherein each of said G1, G2 and Gn is independently
a
cyclic polyamine having a general formula IV:

Image
wherein:
m is an integer from 1 to 10;
n is an integer from 0 to 20;
X and Z are each independently selected from the group consisting of an
oxygen atom, a sulfur atom and a -NH group;
Y1 and Yn are each independently selected from the group consisting of an
oxygen atom, a sulfur atom and a -NH group;




142

A is an alkylene chain having between 1 and 10 substituted and/or non-
substituted carbon atoms;
B1 and Bn are each independently an alkylene chain having between 1 and 20
substituted and/or non-substituted carbon atoms; and
D is a bridging group having a general formula V:
U-W-V
Formula V
whereas:
U and V are each independently selected from the group consisting of
substituted hydrocarbon chain and non-substituted hydrocarbon chain; and
W is selected from the group consisting of amide, ether, ester, disulfide,
thioether, thioester, imine and alkene,
provided that at least one of said X, Z, Y1 and Yn is a -NH group and/or at
least one of said carbon atoms in said alkylene chains is substituted by an
amine
group.

168. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 167, wherein said A is an alkylene chain having a general
formula II:
Image
wherein:
g is an integer that equals 0 or 3-10; and
each of R1, R2 and Rg is independently selected from the group consisting of
hydrogen, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroalicyclic,
heteroaryl, halo,
amino, alkylamino, arylamino, cycloalkylamino, heteroalicyclic amino,
heteroarylamino, hydroxy, alkoxy, aryloxy, azo, C-amido, N-amido, ammonium,
thiohydroxy, thioalkoxy, thioaryloxy, sulfonyl, sulfinyl, N-sulfonamide, S-
sulfonamide phosphonyl, phosphinyl, phosphonium, carbonyl, thiocarbonyl, C-





143

carboxy, O-carboxy, C-thiocarboxy, O-thiocarboxy, N-carbamate, O-carbamate, N-
thiocarbamate, O-thiocarbamate, urea, thiourea, borate, borane, boroaza,
silyl, siloxy,
silaza, aquo, alcohol, peroxo, amine oxide, hydrazine, alkyl hydrazine, aryl
hydrazine,
nitric oxide, cyanate, thiocyanate, isocyanate, isothiocyanate, cyano,
alkylnitrile, aryl
nitrite, alkyl isonitrile, aryl isonitrile, nitrate, nitrite, azido, alkyl
sulfonic acid, aryl
sulfonic acid, alkyl sulfoxide, aryl sulfoxide, alkyl aryl sulfoxide, alkyl
sulfenic acid,
aryl sulfenic acid, alkyl sulfinic acid, aryl sulfinic acid, alkyl thiol
carboxylic acid,
aryl thiol carboxylic acid, alkyl thiol thiocarboxylic acid, aryl thiol
thiocarboxylic
acid, carboxylic acid, alkyl carboxylic acid, aryl carboxylic acid, sulfate,
sulfite,
bisulfate, thiosulfate, thiosulfite, alkyl phosphine, aryl phosphine, alkyl
phosphine
oxide, aryl phosphine oxide, alkyl aryl phosphine oxide, alkyl phosphine
sulfide, aryl
phosphine sulfide, alkyl aryl phosphine sulfide, alkyl phosphonic acid, aryl
phosphonic acid, alkyl phosphinic acid, aryl phosphinic acid, phosphate,
thiophosphate, phosphate, pyrophosphate, triphosphate, hydrogen phosphate,
dihydrogen phosphate, guanidino, S-dithiocarbamate, N-dithiocarbamate,
bicarbonate,
carbonate, perchlorate, chlorate, chlorite, hypochlorite, perbromate, bromate,
bromite,
hypobromite, tetrahalomanganate, tetrafluoroborate, hexafluoroantimonate,
hypophosphite, iodate, periodate, metaborate, tetraarylborate, tetraalkyl
borate,
tartarate, salicylate, succinate, citrate, ascorbate, saccharirate, amino
acid, hydroxamic
acid and thiotosylate.

169. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 168, wherein each of B1 and Bn is independently an
alkylene
chain having a general formula III:
Image
wherein:
p is an integer that equals 0 or g+1;
q is an integer from g+2 to g+20; and




144

each of Rp, Rp+1 and Rq is independently selected from the group consisting
of hydrogen, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroalicyclic,
heteroaryl, halo,
amino, alkylamino, arylamino, cycloalkylamino, heteroalicyclic amino,
heteroarylamino, hydroxy, alkoxy, aryloxy, azo, C-amido, N-amido, ammonium,
thiohydroxy, thioalkoxy, thioaryloxy, sulfonyl, sulfinyl, N-sulfonamide, S-
sulfonamide, phosphonyl, phosphinyl, phosphonium, carbonyl, thiocarbonyl, C-
carboxy, O-carboxy, C-thiocarboxy, O-thiocarboxy, N-carbamate, O-carbamate, N-
thiocarbamate, O-thiocarbamate, urea, thiourea, borate, borane, boroaza,
silyl, siloxy,
silaza, aquo, alcohol, peroxo, amine oxide, hydrazine, alkyl hydrazine, aryl
hydrazine,
nitric oxide, cyanate, thiocyanate, isocyanate, isothiocyanate, cyano,
alkylnitrile, aryl
nitrite, alkyl isonitrile, aryl isonitrile, nitrate, nitrite, azido, alkyl
sulfonic acid, aryl
sulfonic acid, alkyl sulfoxide, aryl sulfoxide, alkyl aryl sulfoxide, alkyl
sulfenic acid,
aryl sulfenic acid, alkyl sulfinic acid, aryl sulfinic acid, alkyl thiol
carboxylic acid,
aryl thiol carboxylic acid, alkyl thiol thiocarboxylic acid, aryl thiol
thiocarboxylic
acid, carboxylic acid, alkyl carboxylic acid, aryl carboxylic acid, sulfate,
sulfite,
bisulfate, thiosulfate, thiosulfite, alkyl phosphine, aryl phosphine, alkyl
phosphine
oxide, aryl phosphine oxide, alkyl aryl phosphine oxide, alkyl phosphine
sulfide, aryl
phosphine sulfide, alkyl aryl phosphine sulfide, alkyl phosphoric acid, aryl
phosphoric acid, alkyl phosphinic acid, aryl phosphinic acid, phosphate,
thiophosphate, phosphate, pyrophosphate, triphosphate, hydrogen phosphate,
dihydrogen phosphate, guanidino, S-dithiocarbamate, N-dithiocarbamate,
bicarbonate,
carbonate, perchlorate, chlorate, chlorite, hypochlorite, perbromate, bromate,
bromite,
hypobromite, tetrahalomanganate, tetrafluoroborate, hexafluoroantimonate,
hypophosphite, iodate, periodate, metaborate, tetraarylborate, tetraalkyl
borate,
tartarate, salicylate, succinate, citrate, ascorbate, saccharirate, amino
acid, hydroxamic
acid and thiotosylate.

170. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 168, wherein at least one of said C1, C2 and Cg is a
chiral carbon
atom.


145

171. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 169, wherein at least one of said Cp, Cp+1 and Cq is a
chiral
carbon atom.
172. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 160, wherein said cyclic polyamine has a general formula
selected from the group consisting of:
Image
wherein:
m is an integer from 1 to 10;
n is an integer from 0 to 20;
X and Z are each independently selected from the group consisting of an
oxygen atom, a sulfur atom and a -NH group;




146
Y, and Yn are each independently selected from the group consisting of an
oxygen atom, a sulfur atom and a -NH group;
A is an alkylene chain having between 1 and 10 substituted and/or non-
substituted carbon atoms;
B1 and Bn are each independently an alkylene chain having between 1 and 20
substituted and/or non-substituted carbon atoms; and
D is a bridging group having a general formula V:
U-W-V
Formula V
whereas:
U and V are each independently selected from the group consisting of
substituted hydrocarbon chain and non-substituted hydrocarbon chain; and
W is selected from the group consisting of amide, ether, ester, disulfide,
thioether, thioester, imine and alkene,
and further wherein should said D is attached at one end to A (Formulas VI,
VII and X), said U or said V are being attached to one carbon atom in said
alkylene
chain and should said D is attached at one end to B1 or Bn (Formulas VIII, IX
and X),
said U or said V are being attached to one carbon atom in said alkylene chain,
provided that at least one of said X, Z, Y1 and Yn is a -NH group and/or at
least one of said carbon atoms in said alkylene chains is substituted by an
amine
group.
173. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 172, wherein said A is an alkylene chain having a general
formula II:
Image
wherein:
g is an integer that equals 0 or 3-10; and


147
each of R1, R2 and Rg is independently selected from the group consisting of
hydrogen, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroalicyclic,
heteroaryl, halo,
amino, alkylamino, arylamino, cycloalkylamino, heteroalicyclic amino,
heteroarylamino, hydroxy, alkoxy, aryloxy, azo, C-amido, N-amido, ammonium,
thiohydroxy, thioalkoxy, thioaryloxy, sulfonyl, sulfinyl, N-sulfonamide, S-
sulfonamide, phosphonyl, phosphinyl, phosphonium, carbonyl, thiocarbonyl, C-
carboxy, O-carboxy, C-thiocarboxy, O-thiocarboxy, N-carbamate, O-carbamate, N-
thiocarbamate, O-thiocarbamate, urea, thiourea, borate, borane, boroaza,
silyl, siloxy,
silaza, aquo, alcohol, peroxo, amine oxide, hydrazine, alkyl hydrazine, aryl
hydrazine,
nitric oxide, cyanate, thiocyanate, isocyanate, isothiocyanate, cyano,
alkylnitrile, aryl
nitrile, alkyl isonitrile, aryl isonitrile, nitrate, nitrile, azido, alkyl
sulfonic acid, aryl
sulfonic acid, alkyl sulfoxide, aryl sulfoxide, alkyl aryl sulfoxide, alkyl
sulfenic acid,
aryl sulfenic acid, alkyl sulfinic acid, aryl sulfinic acid, alkyl thiol
carboxylic acid,
aryl thiol carboxylic acid, alkyl thiol thiocarboxylic acid, aryl thiol
thiocarboxylic
acid, carboxylic acid, alkyl carboxylic acid, aryl carboxylic acid, sulfate,
sulfite,
bisulfate, thiosulfate, thiosulfite, alkyl phosphine, aryl phosphine, alkyl
phosphine
oxide, aryl phosphine oxide, alkyl aryl phosphine oxide, alkyl phosphine
sulfide, aryl
phosphine sulfide, alkyl aryl phosphine sulfide, alkyl phosphonic acid, aryl
phosphonic acid, alkyl phosphinic acid, aryl phosphinic acid, phosphate,
thiophosphate, phosphate, pyrophosphate, triphosphate, hydrogen phosphate,
dihydrogen phosphate, guanidino, S-dithiocarbamate, N-dithiocarbamate,
bicarbonate,
carbonate, perchlorate, chlorate, chlorite, hypochlorite, perbromate, bromate,
bromite,
hypobromite, tetrahalomanganate, tetrafluoroborate, hexafluoroantimonate,
hypophosphite, iodate, periodate, metaborate, tetraarylborate, tetraalkyl
borate,
tartarate, salicylate, succinate, citrate, ascorbate, saccharirate, amino
acid, hydroxamic
acid and thiotosylate.
174. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 173, wherein each of B, and Bn is independently an
alkylene
chain having a general formula III:


148
Image
wherein:
p is an integer that equals 0 or g+1;
q is an integer from g+2 to g+20; and
each of Rp, Rp+1 and Rq is independently selected from the group consisting
of hydrogen, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroalicyclic,
heteroaryl, halo,
amino, alkylamino, arylamino, cycloalkylamino, heteroalicyclic amino,
heteroarylamino, hydroxy, alkoxy, aryloxy, azo, C-amido, N-amido, ammonium,
thiohydroxy, thioalkoxy, thioaryloxy, sulfonyl, sulfinyl, N-sulfonamide, S-
sulfonamide phosphonyl, phosphinyl, phosphonium, carbonyl, thiocarbonyl, C-
carboxy, O-carboxy, C-thiocarboxy, O-thiocarboxy, N-carbamate, O-carbamate, N-
thiocarbamate, O-thiocarbamate, urea, thiourea, borate, borane, boroaza,
silyl, siloxy,
silaza, aquo, alcohol, peroxo, amine oxide, hydrazine, alkyl hydrazine, aryl
hydrazine,
nitric oxide, cyanate, thiocyanate, isocyanate, isothiocyanate, cyano,
alkylnitrile, aryl
nitrite, alkyl isonitrile, aryl isonitrile, nitrate, nitrite, azido, alkyl
sulfonic acid, aryl
sulfonic acid, alkyl sulfoxide, aryl sulfoxide, alkyl aryl sulfoxide, alkyl
sulfenic acid,
aryl sulfenic acid, alkyl sulfinic acid, aryl sulfinic acid, alkyl thiol
carboxylic acid,
aryl thiol carboxylic acid, alkyl thiol thiocarboxylic acid, aryl thiol
thiocarboxylic
acid, alkyl carboxylic acid, carboxylic acid, aryl carboxylic acid, sulfate,
sulfite,
bisulfate, thiosulfate, thiosulfite, alkyl phosphine, aryl phosphine, alkyl
phosphine
oxide, aryl phosphine oxide, alkyl aryl phosphine oxide, alkyl phosphine
sulfide, aryl
phosphine sulfide, alkyl aryl phosphine sulfide, alkyl phosphonic acid, aryl
phosphoric acid, alkyl phosphinic acid, aryl phosphinic acid, phosphate,
thiophosphate, phosphate, pyrophosphate, triphosphate, hydrogen phosphate,
dihydrogen phosphate, guanidino, S-dithiocarbamate, N-dithiocarbamate,
bicarbonate,
carbonate, perchlorate, chlorate, chlorite, hypochlorite, perbromate, bromate,
bromite,
hypobromite, tetrahalomanganate, tetrafluoroborate, hexafluoroantimonate,
hypophosphite, iodate, periodate, metaborate, tetraarylborate, tetraalkyl
borate,



149

tartarate, salicylate, succinate, citrate, ascorbate, saccharirate, amino
acid, hydroxamic
acid and thiotosylate.
175. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 173, wherein at least one of said C1, C2 and Cg is a
chiral carbon
atom.
176. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 174, wherein at least one of said Cp, Cp+1 and Cq is a
chiral
carbon atom.
177. The method, the assay and/or the transplantable hematopoietic cell
preparation of claim 137, wherein said polyamine chelator is selected from the
group
consisting of ethylendiamine, diethylenetriamine, triethylenetetramine,
triethylenediamine, tetraethylenepentamine, aminoethylethanolamine,
aminoethylpiperazine, pentaethylenehexamine, captopril, penicilamine, N,N'-
bis(3-
aminopropyl)-1,3-propanediamine, N,N'-Bis-(2-animoethyl)-1,3-propanediamine,
1,7-dioxa-4,10-diazacyclododecane, 1,4,8,11-tetraaza cyclotetradecane-5,7-
dione,
1,4,7-triazacyclononane, 1-oxa-4,7,10-triazacyclododecane, 1,4,8,12-
tetraazacyclopentadecane, and 1,4,7,10-tetraazacyclododecane.
178. An assay of determining whether a retinoic acid receptor antagonist is
an effective hematopoietic stem cell expansion agent, the assay comprising
culturing
hematopoietic mononuclear cells which comprise a major fraction of
hematopoietic
committed cells and a minor fraction of hematopoietic stem and progenitor
cells in the
presence of the retinoic acid receptor antagonist and monitoring expansion of
said
hematopoietic stem cells, wherein if increased expansion and decreased
differentiation of said hematopoietic stem cells occurs, as compared to non-
treated
hematopoietic mononuclear cells, the retinoic acid receptor antagonist is an
effective
hematopoietic stem cell expansion agent.
179. The assay of claim 178, wherein said retinoic acid receptor antagonist
is selected from the group consisting of: AGN 194310; AGN 193109; 3-(4-Methoxy-




150
phenylsulfanyl)-3-methyl-butyric acid; 6-Methoxy-2,2-dimethyl-thiochroman-4-
one,2,2-Dimethyl-4-oxo-thiochroman-6-yltrifluoromethane-sulfonate; Ethyl 4-
((2,2
dimethyl-4-oxo-thiochroman-6-yl)ethynyl)-benzoate; Ethyl 4-((2,2-dimethy 1-4-
triflouromethanensulfonyloxy -(2H)- thiochromen-6-yl)ethynyl)-benzoate(41);
Thiochromen-6-yl]-ethynyl]-benzoate(yl); (p-[(E)-2-[3'4'-Dihydro-4,4'-dimethyl-
7'-
(heptyloxy)-2'H-1-benzothiopyran-6'yl] propenyl] benzoic acid 1'1'-dioxide;
2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-butoxyphenyl)-3-methyl]-octa-2,4,6-trienoic
acid;
2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-propoxyphenyl)-3-methyl]-octa-2,4,6-trienoic
acid;
2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-pentoxyphenyl)-3-methyl]-octa-2,4,6-trienoic
acid;
2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-hexoxyphenyl)-3-methyl]-octa-2,4,6-trienoic
acid;
2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-heptoxyphenyl)-3-methyl]-octa-2,4,6-trienoic
acid;
2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-octoxyphenyl)-3-methyl]-octa-2,4,6-trienoic
acid;
(2E,4E,6E)-7-[3-t-butyl-5-(1-phenyl-vinyl)-phenyl]-3-methyl-octa-2,4,6-
trienoic acid;
2E,4E,6E-[7-(3,5-Di-t-butyl-4-{[4,5-3 H2]-n-pentoxy}phenyl)-3-
methyl]-
octa-2,4,6-trienoic acid; (2E,4E)-(1RS,2RS)-5-[2-(3,5-di-tert.butyl-2-ethoxy-
phenyl)-
cyclopropyl]-3-methyl-penta-2,4-dienoic acid ethyl ester; (2E,4E)-(1RS,2RS)-5-
[2-
(3,5-di-tert.butyl-2-ethoxy-phenyl)-cyclopropyl]-3-methyl-penta-2,4-dienoic
acid;
(2E,4E)-(1RS,2RS)-5-[2-(3,5-di-tert.butyl-2-butoxy-phenyl)-cyclopropyl]-3-
methyl-
penta-2,4-dienoic acid; (2E,4E,6Z)-7-[3,5-di-tert.butyl-2-ethoxyphenyl]3-
methyl-
2,4,6-octatrienoic acid; (2E,4E,6Z)-7-[3,5-di-tert.butyl-2-butyloxyphenyl]-3-
methyl-
2,4,6-octatrienoic acid; 4-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-
naphthalene-
carboxamido) benzoic acid; (2E,4E)-3-methyl-5-[(1S,2S)-2-(5,5,8,8-tetramethyl-
;
5,6,7,8-tetrahydro-naphthalen-2-yl)-cyclopropyl]-penta-2,4-dienoic acid; p-
[(E)-2-
[3',4'-Dihydro-4',4'-dimethyl-7'-(heptyloxy)-2'H-1-benzothiopyran-6'-
yl]propenyl]benzoic acid; 1',1'-dioxide, 4-(7,7,10,10-Tetramethyl-1-pyridin-3-
ylmethyl-4,5,7,8,9,10-hexahydro-1H-naphto[2,3-g]indol-3-yl)-benzoic acid;
(2E,4E,6Z)-7-[3,5-di-tert.butyl-2-methoxyphenyl]-3-methyl-2,4,6-octatrienoic
acid;
(2E,4E,6Z)-7-[3,5-di-tert.butyl-2-ethoxyphenyl]-3-methyl-2,4,6-octatrienoic
acid;
(2E,4E,6Z)-7-[3,5-di-tert.butyl-2-hexyloxyphenyl]-3-methyl-2,4,6-octatrienoic
acid;
(2E,4E,6Z)-7-[3,5-di-tert.butyl-2-octyloxyphenyl]-3-methyl-2,4,6-octatrienoic
acid;
and (2E,4E)-(1RS,2RS)-5-[2-(3,5-di-tert-butyl-2-butoxy-phenyl)-cyclopropyl]-3-
methyl-penta-2,4-dienoic acid, (2E,4E,6Z)-7-(3-n-propoxy-5,6,7,8-tetrahydro-
5,5,8,8-
tetramethylnaphthalene-2-yl)-3-methylocta-2,4,6-trienoic acid, 4-(5H-2,3(2,5




151
dimethyl-2,5-hexano)-5-n-propyldibenzo[b,e][1,4]diazepin-11-yl)benzoic acid, 4-

(5H-2,3-(2,5-dimethyl-2,5-hexano)-5methyl-8-nitrodibenzo[b,e] [1,4]diazepin-11-

yl)benzoic acid, 4-{[4-(4-Ethylphenyl)2,2-dimethyl-(2H)-thiochromen-6-
yl]ethynyl}benzoic acid, 4-[4-2methyl-1,2-dicarba-closo-dodecaboran-1-yl-
phenylcarbamoyl]benzoic acid, 4-[4,5,7,8,9,10-hexahydro-7,7,10,10-tetramethyl-
1-(3-
pyridylmethyl)-anthra[1,2-b]pyrrol-3-yl]benzoic acid, (3-pyridylmethyl)-]5-
thiaanthra[2,1-b]pyrrol-3-yl)benzoic acid, and (3-pyridylmethyl)-anthra[2ml-
d]pyrazol-3-yl]benzoic acid.
180. An assay of determining whether a retinoid X receptor antagonist is an
effective hematopoietic stem cell expansion agent, the assay comprising
culturing
hematopoietic mononuclear cells which comprise a major fraction of
hematopoietic
committed cells and a minor fraction of hematopoietic stem and progenitor
cells in the
presence of the retinoid X receptor antagonist and monitoring expansion of
said
hematopoietic stem cells, wherein if increased expansion and decreased
differentiation of said hematopoietic stem cells occurs, as compared to non-
treated
hematopoietic mononuclear cells, the retinoid X receptor antagonist is an
effective
hematopoietic stem cell expansion agent.
181. The assay of claim 180, wherein said retinoid X receptor antagonist is
selected from the group consisting of LGN100572, LGN100574, 1-(3-hydroxy-
5,6,7,8-tetrahydro-5,5,8,8-tetramethylnaphthalene-2-yl)ethanone, 1-(3-propoxy-
5,6,7,8-tetrahydro-5,5,8,8-tetramethylnaphthalene-2-yl)ethanone, 3-(3-propoxy-
5,6,7,8-tetrahydro-5,5,8,8-tetramethylnaphthalene-2-yl)but-2-enenitrile, 3-(3-
propoxy-5,6,7,8-tetrahydro-5,5,8,8-tetramethylnaphthalene-2-yl)but-2-enal,
(2E,4E,6E)-7-3[-propoxy-5,6,7,8-tetrahydro 5,5,8,8-tetramethyl-2-naphthalene-2-
yl]-
3-methylocta-2,4,6-trienoic acid, 4-[3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-
2-
naphthyl)carbonyl] benzoic acid, 4-[1-(3,5, 5,8,8-pentamethyl-5,6,7,8-
tetrahydro-2-
naphthyl)ethenyl] benzoic acid, 4-[1(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-
2-
naphthyl)cyclopropyl] benzoic acid, 4-[1-(3,5,5,8,8-pentamethyl-5,6,7,8-
tetrahydro-2-
naphthyl)ethenyl] benzenete trazole, 2-[1-(5,5,8,8-tetramethyl-5,6,7,8-
tetrahydro-2-
naphthyl) ethenyl]pyridine-5-carboxylic acid, 2-[1-(3,5,5,8,8-pentamethyl-
5,6,7,8-
tetrahydro-2-naphthyl)ethyl]pyridine-5-carboxylic acid, ethyl-2-[1-(3,5,5,8, 8-




152
pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)ethenyl]pyridine-5-carboxylate, 5-[1-

3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)ethenyl]pyridine-2-
carboxylic
acid, 2-[1-(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)
cyclopropyl]pyridine-5-carboxylic acid, methyl 2-[1-(3,5,5,8,8-pentamethyl-
5,6,7,8-
tetrahydro-2-naphthyl)cyclopropyl]pyridine-5-carboxylate, 4-[1-(3,5, 5,8,8-
pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)ethenyl]-N-(4-hydroxyphenyl)
benzamide,
2-[1-(3,5,5,8,8-Pentamethyl-5,6,7,8-tetrahydro-2-naphthyl) ethenyl] pyridine-5-

carboxylic acid, 2-[1-(3,5,5,8,8-Pentamethyl-5, 6,7,8-tetrahydro-2-
naphthyl)cyclopropyl]pyridine-5-carboxylic acid, 4-[(3,5, 5,8,8-pentamethyl-
5,6,7,8-
tetrahydro-2-naphthyl)carbonyl]benzoic acid butyloxime, 4-[(3,5,5,8,8-
pentamethyl-
5,6,7,8-tetrahydro-2-naphthyl) carbonyl]benzoic acid propyloxime, 4-
[(3,5,5,8,8-
pentamethyl-5,6,7,8-terrahydro-2-naphthyl)carbonyl]benzoic acid cyanoimine, 4-
[(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)carbonyl]benzoic acid
allyloxime, 4-[(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-
naphthyl)carbonyl]benzoic
acid 4-(3-methylbut-2-enoic acid)oxime, and 4-[(3,5,5,8,8-pentamethyl-5,6,7,8-
tetrahydro-2-naphthyl)carbonyl]benzoic acid 1-aminoethyloxime, (2E,4E,6Z)-7-(3-
n-
propoxy-5,6,7,8-tetrahydro-5,5,8,8-tetramethylnaphthalene-2-yl)-3-methylocta-
2,4,6-
trienoic acid, 4-(5H-2,3(2,5 dimethyl-2,5-hexano)-5-n-
propyldibenzo[b,e][1,4]diazepin-11-yl)benzoic acid, and 4-(5H-2,3-(2,5-
dimethyl-
2,5-hexano)-5methyl-8-nitrodibenzo[b,e][1,4]diazepin-11-yl)benzoic acid.
182. An assay of determining whether a vitamin D receptor antagonist is an
effective hematopoietic stem cell expansion agent, the assay comprising
culturing
hematopoietic mononuclear cells which comprise a major fraction of
hematopoietic
committed cells and a minor fraction of hematopoietic stem and progenitor
cells in the
presence of the vitamin D receptor antagonist and monitoring expansion of said
hematopoietic stem cells, wherein if increased expansion and decreased
differentiation of said hematopoietic stem cells occurs, as compared to non-
treated
hematopoietic mononuclear cells, the vitamin D receptor antagonist is an
effective
hematopoietic stem cell expansion agent.
183. The assay of claim 182, wherein said Vitamin D receptor antagonist is
selected from the group consisting of: 1 alpha, 25-(OH)-D3-26,23 lactone; 1
alpha, 25-


153
dihydroxyvitamin D (3); the 25-carboxylic ester ZK159222; (23S)- 25-dehydro-1
alpha-OH-D (3); (23R)-25-dehydro-1 alpha-OH-D (3); 1 beta, 25 (OH)2 D3; 1
beta,
25(OH)2-3-epi-D3; (23S) 25-dehydro-1 alpha(OH) D3-26,23-lactone; (23R) 25-
dehydro-1 alpha(OH)D3-26,23-lactone and Butyl-(5Z,7E,22E-(1S,7E,22E-
(1S,3R,24R)-1,3,24-trihydroxy-26,27-cyclo-9,10-secocholesta-5,7,10(19),22-
tetraene-
25-carboxylate).
184. An assay of determining whether an agent that inhibits PI 3-kinase
activity is an effective hematopoietic stem cell expansion agent, the assay
comprising
culturing hematopoietic mononuclear cells which comprise a major fraction of
hematopoietic committed cells and a minor fraction of hematopoietic stem and
progenitor cells in the presence of the agent that inhibits PI 3-kinase
activity and
monitoring expansion of said hematopoietic stem cells, wherein if increased
expansion and decreased differentiation of said hematopoietic stem cells
occurs, as
compared to non-treated hematopoietic mononuclear cells, the agent that
inhibits PI 3-
kinase activity is an effective hematopoietic stem cell expansion agent.
185. An assay of determining whether a nicotinamide analog, a
nicotinamide or a nicotinamide analog derivative or a nicotinamide or a
nicotinamide
analog metabolite is an effective hematopoietic stem cell expansion agent, the
assay
comprising culturing hematopoietic mononuclear cells which comprise a major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells in the presence of a nicotinamide analog, a nicotinamide
or a
nicotinamide analog derivative or a nicotinamide or a nicotinamide analog
metabolite
and monitoring expansion of said hematopoietic stem cells, wherein if
increased
expansion and decreased differentiation of said hematopoietic stem cells
occurs, as
compared to non-treated hematopoietic mononuclear cells, the a nicotinamide
analog,
a nicotinamide or a nicotinamide analog derivative or a nicotinamide or a
nicotinamide analog metabolite is an effective hematopoietic stem cell
expansion
agent.



154
186. The assay of any of claims 178, 180, 182, 184 and 185, wherein
culturing said hematopoietic mononuclear cells is performed in a presence of
an
effective amount of a cytokine.
187. The assay of claim 186, wherein said cytokine is an early acting
cytokines.
188. The assay of claim 187, wherein said early acting cytokine is selected
from the group comprising stem cell factor, FLT3 ligand, interleukin-1,
interleukin-2,
interleukin-3, interleukin-6, interleukin-10, interleukin-12, tumor necrosis
factor-a
and thrombopoietin.
189. The assay of claim 186, wherein said cytokine is a late acting
cytokines.
190. The assay of claim 189, wherein said late acting cytokines are selected
from the group comprising granulocyte colony stimulating factor,
granulocyte/macrophage colony stimulating factor, erythropoietin, FGF, EGF,
NGF,
VEGF, LIF, Hepatocyte growth factor and macrophage colony stimulating factor.
191. The assay of any of claims 178, 180, 182, 184 and 185, wherein said
hematopoietic mononuclear cells are derived from a source selected from the
group
consisting of bone marrow, peripheral blood and neonatal umbilical cord blood.
192. The assay of any of claims 178, 180, 182, 184 and 185, wherein said
monitoring decreased differentiation is by determining hematopoietic cell
surface
expression of CD34.
193. The assay of any of claims 178, 180, 182, 184 and 185, wherein said
monitoring decreased differentiation is by determining an absence, or
significantly
diminished hematopoietic cell surface expression of CD38, CD3, CD61, CD19,
CD33, CD14, CD15 or CD4.


155

194. A hematopoietic stem cells collection/culturing bag supplemented with
an effective amount of a retinoic acid receptor antagonist, a retinoid X
receptor
antagonist and/or a Vitamin D receptor antagonist, which substantially
inhibits cell
differentiation of a hematopoietic stem cells fraction of hematopoietic
mononuclear
cells which comprise a major fraction of hematopoietic committed cells and a
minor
fraction of hematopoietic stem and progenitor cells.
195. The hematopoietic stem cells collection/culturing bag of claim 194,
wherein said retinoic acid receptor antagonist is selected from the group
consisting of
AGN 194310; AGN 193109; 3-(4-Methoxy-phenylsulfanyl)-3-methyl-butyric acid;
6-Methoxy-2,2-dimethvl-thiochroman-4-one,2,2-Dimethyl-4-oxo-thiochroman-6-
yltrifluoromethane-sulfonate; Ethyl 4-((2,2 dimethyl-4-oxo-thiochroman-6-
yl)ethynyl)-benzoate; Ethyl 4-((2,2-dimethy 1-4-triflouromethanensulfonyloxy -
(2H)-
thiochromen-6-yl)ethynyl)-benzoate(41); Thiochromen-6-yl]-ethynyl]-
benzoate(yl);
(p-[(E)-2-[3'4'-Dihydro-4,4'-dimethyl-7'-(heptyloxy)-2'H-1-benzothiopyran-
6'yl]
propenyl] benzoic acid 1'1'-dioxide; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
butoxyphenyl)-
3-methyl]-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
propoxyphenyl)-
3-methyl]-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
pentoxyphenyl)-
3-methyl]-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
hexoxyphenyl)-3-
methyl]-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
heptoxyphenyl)-3-
methyl]-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
octoxyphenyl)-3-
methyl]-octa-2,4,6-trienoic acid; (2E,4E,6E)-7-[3-t-butyl-5-(1-phenyl-vinyl)-
phenyl]-
3-methyl-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-{[4,5-3
H2]-n-pentoxy}phenyl)-3-methyl]-octa-2,4,6-trienoic acid; (2E,4E)-
(1RS,2RS)-
S-[2-(3,S-di-tert.butyl-2-ethoxy-phenyl)-cyclopropyl]-3-methyl-penta-2,4-
dienoic acid
ethyl ester; (2E,4E)-(1RS,2RS)-S-[2-(3,5-di-tert.butyl-2-ethoxy-phenyl)-
cyclopropyl]-
3-methyl-penta-2,4-dienoic acid; (2E,4E)-(1RS,2RS)-S-[2-(3,5-di-tert.butyl-2-
butoxy-phenyl)-cyclopropyl]-3-methyl-penta-2,4-dienoic acid; (2E,4E,6Z)-7-[3,5-
di-
tert.butyl-2-ethoxyphenyl]3-methyl-2,4,6-octatrienoic acid; (2E,4E,6Z)-7-[3,5-
di-
tert.butyl-2-butyloxyphenyl]-3-methyl-2,4,6-octatrienoic acid; 4-(5,6,7,8-
tetrahydro-
5,5,8,8-tetramethyl-2-naphthalene-carboxamido) benzoic acid; (2E,4E)-3-methyl-
5-
[(1S,2S)-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydro-naphthalen-2-yl)-
cyclopropyl]-
penta-2,4-dienoic acid; p-[(E)-2-[3',4'-Dihydro-4',4'-dimethyl-7'-(heptyloxy)-
2'H-1-


156
benzothiopyran-6'-yl]propenyl]benzoic acid; 1',1'-dioxide, 4-(7,7,10,10-
Tetramethyl-
1-pyridin-3-ylmethyl-4,5,7,8,9,10-hexahydro-1H-naphto[2,3-g]indol-3-yl)-
benzoic
acid; (2E,4E,6Z)-7-[3,5-di-tert.butyl-2-methoxyphenyl]-3-methyl-2,4,6-
octatrienoic
acid; (2E,4E,6Z)-7-[3,5-di-tert.butyl-2-ethoxyphenyl]-3-methyl-2,4,6-
octatrienoic
acid; (2E,4E,6Z)-7-[3,5-di-tert.butyl-2-hexyloxyphenyl]-3-methyl-2,4,6-
octatrienoic
acid; (2E,4E,6Z)-7-[3,5-di-tert.butyl-2-octyloxyphenyl]-3-methyl-2,4,6-
octatrienoic
acid; and (2E,4E)-(1RS,2RS)-5-[2-(3,5-di-tert-butyl-2-butoxy-phenyl)-
cyclopropyl]-
3-methyl-penta-2,4-dienoic acid. (2E,4E,6Z)-7-(3-n-propoxy-5,6,7,8-tetrahydro-
5,5,8,8-tetramethylnaphthalene-2-yl)-3-methylocta-2,4,6-trienoic acid, 4-(5H-
2,3(2,5
dimethyl-2,5-hexano)-5-n-propyldibenzo[b,e] [1,4]diazepin-11-yl)benzoic acid, 4-

(5H-2,3-(2,5-dimethyl-2,5-hexano)-5methyl-8-nitrodibenzo[b,e][1,4]diazepin-11-
yl)benzoic acid, 4-{[4-(4-Ethylphenyl)2,2-dimethyl-(2H)-thiochromen-6-
yl]ethynyl}benzoic acid, 4-[4-2methyl-1,2-dicarba-closo-dodecaboran-1-yl-
phenylcarbamoyl]benzoic acid, 4-[4,5,7,8,9,10-hexahydro-7,7,10,10-tetramethyl-
1-(3-
pyridylmethyl)-anthra[1,2-b]pyrrol-3-yl]benzoic acid, (3-pyridylmethyl)-]5-
thiaanthra[2,1-b]pyrrol-3-yl)benzoic acid, and (3-pyridylmethyl)-anthra[2m1-
d]pyrazol-3-yl]benzoic acid.
196. The hematopoietic stem cell collection bag of claim 194, wherein said
retinoid X receptor antagonist is selected from the group consisting of:
LGN100572,
LGN100574, 1-(3-hydroxy-5,6,7,8-tetrahydro-5,5,8,8-tetramethylnaphthalene-2-
yl)ethanone, 1-(3-propoxy-5,6,7,8-tetrahydro-5,5,8,8-tetramethylnaphthalene-2-
yl)ethanone, 3-(3-propoxy-5,6,7,8-tetrahydro-5,5,8,8-tetramethylnaphthalene-2-
yl)but-2-enenitrile, 3-(3-propoxy-5,6,7,8-tetrahydro-5,5,8,8-
tetramethylnaphthalene-
2-yl)but-2-enal, (2E,4E,6E)-7-3[-propoxy-5,6,7,8-tetrahydro 5,5,8,8-
tetramethyl-2-
naphthalene-2-yl]-3-methylocta-2,4,6-trienoic acid, 4-[3,5,5,8,8-pentamethyl-
5,6,7,8-
tetrahydro-2-naphthyl)carbonyl] benzoic acid, 4-[1-(3,5, 5,8,8-pentamethyl-
5,6,7,8-
tetrahydro-2-naphthyl)ethenyl] benzoic acid, 4-[1(3,5,5,8,8-pentamethyl-
5,6,7,8-
tetrahydro-2-naphthyl)cyclopropyl] benzoic acid, 4-[1-(3,5,5,8,8-pentamethyl-
5,6,7,8-
tetrahydro-2-naphthyl)ethenyl] benzenete trazole, 2-[1-(5,5,8,8-tetramethyl-
5,6,7,8-
tetrahydro-2-naphthyl) ethenyl]pyridine-5-carboxylic acid, 2-[1-(3,5,5,8,8-
pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)ethyl]pyridine-5-carboxylic acid,
ethyl-2-
[1-(3,5,5,8, 8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)ethenyl]pyridine-5-


157
carboxylate, 5-[1-3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-
naphthyl)ethenyl)pyridine-2-carboxylic acid, 2-[1-(3,5,5,8,8-pentamethyl-
5,6,7,8-
tetrahydro-2-naphthyl) cyclopropyl]pyridine-5-carboxylic acid, methyl 2-[1-
(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)cyclopropyl]pyridine-5-
carboxylate, 4-[1-(3,5, 5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-
naphthyl)ethenyl]-N-
(4-hydroxyphenyl) benzamide, 2-[1-(3,5,5,8,8-Pentamethyl-5,6,7,8-tetrahydro-2-
naphthyl) ethenyl] pyridine-5-carboxylic acid, 2-[1-(3,5,5,8,8-Pentamethyl-5,
6,7,8-
tetrahydro-2-naphthyl)cyclopropyl]pyridine-5-carboxylic acid, 4-[(3,5, 5,8,8-
pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)carbonyl]benzoic acid butyloxime, 4-
[(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl) carbonyl]benzoic acid
propyloxime, 4-[(3,5,5,8,8-pentamethyl-5,6,7,8-terrahydro-2-
naphthyl)carbonyl]benzoic acid cyanoimine, 4-[(3,5,5,8,8-pentamethyl-5,6,7,8-
tetrahydro-2-naphthyl)carbonyl]benzoic acid allyloxime, 4-[(3,5,5,8,8-
pentamethyl-
5,6,7,8-tetrahydro-2-naphthyl)carbonyl]benzoic acid 4-(3-methylbut-2-enoic
acid)oxime, and 4-[(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-
naphthyl)carbonyl)benzoic acid 1-aminoethyloxime, (2E,4E,6Z)-7-(3-n-propoxy-
5,6,7,8-tetrahydro-5,5,8,8-tetramethylnaphthalene-2-yl)-3-methylocta-2,4,6-
trienoic
acid, 4-(5H-2,3(2,5 dimethyl-2,5-hexano)-5-n-propyldibenzo[b,e][1,4]diazepin-
11-
yl)benzoic acid, and 4-(5H-2,3-(2,5-dimethyl-2,5-hexano)-5methyl-8-
nitrodibenzo[b,e][1,4]diazepin-11-yl)benzoic acid.
197. The hematopoietic stem cell collection bag of claim 194, wherein said
Vitamin D receptor antagonist is selected from the group consisting of 1
alpha, 25-
(OH)-D3-26,23 lactone; 1 alpha, 25-dihydroxyvitamin D (3); the 25-carboxylic
ester
ZK159222; (235)- 25-dehydro-1 alpha-OH-D (3); (23R)-25-dehydro-1 alpha-OH-D
(3); 1 beta, 25 (OH)2 D3; 1 beta, 25(OH)2-3-epi-D3; (235) 25-dehydro-1
alpha(OH)
D3-26,23-lactone; (23R) 25-dehydro-1 alpha(OH)D3-26,23-lactone and Butyl-
(5Z,7E,22E-(1S,7E,22E-(1S,3R,24R)-1,3,24-trihydroxy-26,27-cyclo-9,10-
secocholesta-5,7,10(19),22-tetraene-25-carboxylate).
198. A hematopoietic stem cells collection/culturing bag supplemented with
an effective amount of nicotinamide, a nicotinamide analog, a nicotinamide or
a
nicotinamide analog derivative or a nicotinamide or a nicotinamide analog
metabolite,



158
which substantially inhibits cell differentiation of a hematopoietic stem
cells fraction
of hematopoietic mononuclear cells which comprise a major fraction of
hematopoietic
committed cells and a minor fraction of hematopoietic stem and progenitor
cells.
199. A hematopoietic stem cells collection/culturing bag supplemented with
an effective amount of an agent that inhibits PI 3-kinase activity, which
substantially
inhibits cell differentiation of a hematopoietic stem cells fraction of
hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells.
200. An ex-vivo expanded population of hematopoietic stem cells, obtained
by the method of any of claims 1, 38, 59, 80, 95 and 118.

Description

Note: Descriptions are shown in the official language in which they were submitted.




CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
EX VIVO EXPANSION OF HEMATOPOIETIC STEM CELL POPULATIONS IN
MONONUCLEAR CELL CULTURES
FIELD AND BACKGROUND OF THE INVENTION
The present invention relates to methods of ex-vivo expansion (self renewal)
of hematopoietic stem cells present in the hematopoietic mononuclear cells
fraction of
a blood sample and to expanded (self renewed) populations of hematopoietic
stem
cells obtained thereby. The present invention further relates to therapeutic
applications in which these methods and/or the expanded hematopoietic stem
cell
populations obtained thereby are utilized.
An increasing need for ex-vivo cultures of hematopoietic stem cells has
arisen,
in particular for purposes such as stem cell expansion and retroviral-mediated
gene
transduction. Methods for generating ex-vivo cultures of stem cells, however,
typically result in a rapid decline in stem cell population activity, further
resulting in a
markedly impaired self renewal potential and diminished transplantability of
the
cultured cell populations. The need to improve such methods is widely
acknowledged. Additionally, applications in gene therapy using retroviral
vectors
necessitate the use of proliferating hematopoietic stem cells, yet require
that these
cells remain undifferentiated while in culture, in order to maintain long-term
expression of the transduced gene. Thus, the ability to maintain ex-vivo
cultures of
hematopoietic stem cell populations with long-term, self renewal capacity is
of
critical importance for a wide array of medical therapeutic applications.
Presently, expansion of renewable stem cells have been achieved either by
growing the stem cells over a feeder layer of fibroblast cells, or by growing
the cells
in the presence of the early acting cytokines thrombopoietin (TPO),
interleukin-6 (IL
6), an FLT-3 ligand and stem cell factor (SCF) (Madlambayan GJ et al. (2001) J
Hematother Stem Cell Res 10: 481, Punzel M et al. (1999) Leukemia 13: 92, and
Lange W et al. (1996) Leukemia 10: 943). While expanding stem cells over a
feeder
layer results in vast, substantially endless cell expansion, expanding stem
cells
without a feeder layer, in the presence of the early acting cytokines listed
above,
results in an elevated degree of differentiation (see controls described in
the Examples
section and Leslie NR et al. (Blood (1998) 92: 4798), Petzer AL et al. (1996)
J Exp
Med Jun 183: 2551, Kawa Y et al. (2000) Pigment Cell Res 8: 73).



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
2
Hence, self renewal (expansion) of hemopoietic stem and progenitor cells,
both in vivo and in vitro, is limited by cell differentiation. Differentiation
in the
hematopoietic system involves, among other changes, altered expression of
surface
antigens (Sieff C, Bicknell D, Caine G, Robinson J, Lam G, Greaves MF (1982)
Changes in cell surface antigen expression during hematopoietic
differentiation.
Blood 60:703). In normal human, most of the hematopoietic pluripotent stem
cells
and the lineage committed progenitor cells are CD34+. The majority of cells
are
CD34+CD38+, with a minority of cells (< 10 %) being CD34+CD38-. The
CD34+CD38- phenotype appears to identify the most immature hematopoietic
cells,
1o which are capable of self renewal and multilineage differentiation. The
CD34+CD38- cell fraction contains more long-term culture initiating cells (LTC-
IC)
pre-CFU and exhibits longer maintenance of their phenotype and delayed
proliferative
response to cytokines as compared with CD34+CD38+ cells. CD34+CD38- cells can
give rise to lymphoid and myeloid cells in vitro and have an enhanced capacity
to
~5 repopulate SCID mice (Bhatia M, Wang JCY, Kapp U, Bonnet D, Dick JE (1997)
Purification of primitive human hematopoietic cells capable of repopulating
immune-
deficient mice. Proc Natl Acad Sci USA 94:5320). Moreover, in patients who
received autologous blood cell transplantation, the number of CD34+CD38- cells
infused correlates positively with the speed of hematopoietic recovery. In
line with
2o these functional features, CD34+CD38- cells have been shown to have
detectable
levels of telomerase.
The presently published works on ex-vivo expansion of hematopoietic stem
and progenitor cells involve starting inoculums of cells, which are highly
enriched
with progenitor cells that express CD34 or, the even earlier, AC133 antigens
[Dexter,
25 T.M., T.D. Allen, and L.G. Lajtha, Conditions controlling the proliferation
of
haemopoietic stem cells in vitro. J.Cell Physiol, 1977. 91(3): p. 335-44;
Muench,
M.O., J.G. Schneider, and M.A. Moore, Interactions among colony-stimulating
factors, IL-1 beta, IL-6, and kit-ligand in the regulation of primitive murine
hematopoietic cells. Exp. Hematol., 1992. 20(3): p. 339-49; Verfaillie, C.M.,
Direct
30 contact between human primitive hematopoietic progenitors and bone marrow
stroma
is not required for long-term in vitro hematopoiesis. Blood, 1992. 79(11): p.
2821-26;
Migliaccio, G., A.R. Migliaccio, M.L. Druzin, P.J. Giardina, K.M. Zsebo, and
J.W.
Adamson, Long-term generation of colony-forming cells in liquid culture of
CD34+



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
3
cord blood cells in the presence of recombinant human stem cell factor. Blood,
1992.
79(10): p. 2620-27; Purdy, M.H., C.J. Hogan, L. Hami, I. McNiece, W. Franklin,
R.B.
Jones, S.I. Bearman, R.J. Berenson, P.J. Cagnoni, and S. Heimfeld, Large
volume ex-
vivo expansion of CD34-positive hematopoietic progenitor cells for
transplantation. J.
Hematother., 1995. 4(6): p. 515-25; McNiece, L, R. Andrews, M. Stewart, S.
Clark,
T. Boone, and P. Quesenberry, Action of interleukin-3, G-CSF, and GM-CSF on
highly enriched human hematopoietic progenitor cells: synergistic interaction
of GM-
CSF plus G-CSF. Blood, 1989. 74(1): p. 110-14; Colter, M., M. Jones, and S.
Heimfeld, CD34+ progenitor cell selection: clinical transplantation, tumor
cell
purging, gene therapy, ex-vivo expansion, and cord blood processing. J
Hematother,
1996. 5(2): p. 179-84; Kohler, T., R. Plettig, W. Wetzstein, B. Schaffer, R.
Ordemann,
H.O. Nagels, G. Ehninger, and M. Bornhauser, Defining optimum conditions for
the
ex-vivo expansion of human umbilical cord blood cells. Influences of
progenitor
enrichment, interference with feeder layers, early-acting cytokines and
agitation of
culture vessels. Stem Cells, 1999. 17(1): p. 19-24].
As it was shown that initiation of ex-vivo cultures with the entire
mononuclear
cells (MNC) fraction in the presence of cytokines led to expansion of CFUc
during
the first weeks of culturing, followed by a rapid deterioration of the
cultures, it has
been widely accepted heretofore that purification of CD34+ (or AC 133) cells
is a
prerequisite for achieving successful ex-vivo expansion of hematopoietic stem
cells
[Briddell, R., Keren, B.P., Zilm, K.L., et al. Purification of CD34+ cell is
essential for
optimal ex-vivo expansion of umbilical cord blood cells. J. Hematother. 6:145,
1997;
Ian K McNiece, Gregory B. Stoney, Brent P. Keren, and Robert A. Briddell CD34+
cell selection from frozen cord blood products using Isolex 300i and
cliniMACSTM
selection device. Journal of hematotherapy 7:457-461 (1998)] as well as long-
term
culture colony forming cells (LTC-CFUc).
WO 99/40783, WO 00/18885 and Peled et al, Brit. J. Haematol. 116:655 2002,
all of which are incorporated by reference as if fully set forth herein, teach
the effect
of free copper present in cells on the modulation of the balance between self
renewal
and differentiation of hematopoietic progenitor cells. These references teach
that the
addition of agents that are capable of reducing the cell copper content, along
with
early acting cytokines, to CD34+ cell cultures results in long term CD34+ cell
expansion ex-vivo in culture. According to teachings of these references, such
agents



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
4
preferably include transition metal chelators that are capable of binding
copper, such
as, for example, linear polyamines (e.g., tetraethylenepentamine, TEPA).
Hence, it is
shown in these references that the addition of 5-10 ~M TEPA to CD34+ cell
cultures
in the presence of early acting cytokines reduced cell copper content by 30 %
(as
s measured by atomic absorption), and extended the duration of the long-term
cultures
in terms of long-term CFU and CD34+ cell expansion.
However, the methods disclosed in these references also involve purification
of stem or progenitor cells prior to their expansion in cultures.
Thus, using present day technology, stem cells cannot be expanded unless first
substantially enriched or isolated to homogeneity and therefore the presently
known
methods of ex-vivo expanding stem cell populations are limited by the
laborious and
costly process of stem cells enrichment prior to initiation of cultures.
There is thus a widely recognized need for, and it would be highly
advantageous to have, methods of ex-vivo expanding hematopoietic stem cells
without
prior stem cells enrichment.
SUMMARY OF THE INVENTION
The present invention discloses the use of various agents in expanding
hematopoietic stem cells present in the hematopoietic mononuclear cells
fraction of a
blood sample, without the use of a prior stem cells enrichment procedure, to
expanded
(self renewed) populations of hematopoietic stem cells obtained thereby and to
their
uses.
According to one aspect of the present invention there is provided a method of
ex-vivo expanding a population of hematopoietic stem cells, while at the same
time,
2s substantially inhibiting differentiation of the hematopoietic stem cells ex-
vivo.
In one embodiment, the method comprises providing hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells, with ex-vivo
culture
conditions for ex-vivo cell proliferation and, at the same time, for reducing
an
expression and/or activity of CD38, thereby expanding a population of the
hematopoietic stem cells, while at the same time, substantially inhibiting
differentiation of the hematopoietic stem cells ex-vivo.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
In another embodiment the method comprises providing the hematopoietic
mononuclear cells with ex-vivo culture conditions for ex-vivo cell
proliferation and, at
the same time, for reducing a capacity of the hematopoietic mononuclear cells
in
responding to retinoic acid, retinoids and/or Vitamin D, thereby expanding the
5 population of the hematopoietic stem cells while at the same time,
substantially
inhibiting differentiation of the stem cells ex-vivo.
In still another embodiment the method comprises providing the hematopoietic
mononuclear cells with ex-vivo culture conditions for ex-vivo cell
proliferation and, at
the same time, for reducing a capacity of the hematopoietic mononuclear cells
in
responding to signaling pathways involving the retinoic acid receptor,
retinoid-X
receptor and/or Vitamin D receptor, thereby expanding the population of the
hematopoietic stem cells while at the same time, substantially inhibiting
differentiation of the hematopoietic stem cells ex-vivo.
In yet another embodiment the method comprises providing the hematopoietic
mononuclear cells with ex-vivo culture conditions for ex-vivo cell
proliferation and, at
the same time, for reducing a capacity of the hematopoietic mononuclear cells
in
responding to signaling pathways involving PI 3-kinase, thereby expanding the
population of the hematopoietic stem cells while at the same time,
substantially ,
inhibiting differentiation of the hematopoietic stem cells ex-vivo.
In still another embodiment, the method comprises providing the
hematopoietic mononuclear cells with ex-vivo culture conditions for ex-vivo
cell
proliferation and, at the same time, with nicotinamide, a nicotinamide analog,
a
nicotinamide or a nicotinamide analog derivative or a nicotinamide or a
nicotinamide
analog metabolite, thereby expanding the population of the hematopoietic stem
cells
while at the same time, substantially inhibiting differentiation of the
hematopoietic
stem cells ex-vivo.
In yet another embodiment, the method comprises providing the hematopoietic
mononuclear cells with ex-vivo culture conditions for ex-vivo cell
proliferation and, at
the same time, with a PI 3-kinase inhibitor, thereby expanding the population
of the
hematopoietic stem cells while at the same time, substantially inhibiting
differentiation of the hematopoietic stem cells ex-vivo.
In still another embodiment, the method comprises providing the
hematopoietic mononuclear cells with ex-vivo culture conditions for ex-vivo
cell



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
6
proliferation and, at the same time, with one or more copper chelator(s) or
copper
chelate(s), thereby expanding the population of the hematopoietic stem cells
while at
the same time, substantially inhibiting differentiation of the hematopoietic
stem cells
ex-vivo.
Further according to the present invention, there are provided ex-vivo
expanded populations of hematopoietic stem cells, obtained by the methods
described
hereinabove.
According to another aspect of the present invention there is provided a
method of hematopoietic cells transplantation or implantation.
In one embodiment, the method comprises (a) obtaining hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells from a donor;
(b)
providing the hematopoietic mononuclear cells with ex-vivo culture conditions
for cell
proliferation and, at the same time, for reducing an expression and/or
activity of
CD38, thereby expanding a population of the hematopoietic stem cells, while at
the
same time, substantially inhibiting differentiation of the hematopoietic stem
cells ex-
vivo; and (c) transplanting or implanting the hematopoietic stem cells to a
recipient.
In another embodiment, the method comprises (a) obtaining hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
2o and a minor fraction of hematopoietic stem and progenitor cells from a
donor; (b)
providing the hematopoietic mononuclear cells with ex-vivo culture conditions
for cell
proliferation and, at the same time, for reducing a capacity of the
hematopoietic
mononuclear cells in responding to retinoic acid, retinoids and/or Vitamin D,
thereby
expanding a population of the hematopoietic stem cells, while at the same
time,
substantially inhibiting differentiation of the hematopoietic stem cells ex-
vivo; and (c)
transplanting or implanting the hematopoietic stem cells to a recipient.
In yet another embodiment, the method comprises (a) obtaining hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells from a donor;
(b)
3o providing the hematopoietic mononuclear cells with ex-vivo culture
conditions for cell
proliferation and, at the same time, for reducing a capacity of the
hematopoietic
mononuclear cells in responding to responding to signaling pathways involving
the
retinoic acid receptor, the retinoid X receptor and/or the Vitamin D receptor,
thereby



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
7
expanding a population of the hematopoietic stem cells, while at the same
time,
substantially inhibiting differentiation of the hematopoietic stem cells ex-
vivo; and (c)
transplanting or implanting the hematopoietic stem cells to a recipient.
In still another embodiment, the method comprises (a) obtaining
hematopoietic mononuclear cells which comprise a major fraction of
hematopoietic
committed cells and a minor fraction of hematopoietic stem and progenitor
cells from
a donor; (b) providing the hematopoietic mononuclear cells with ex-vivo
culture
conditions for cell proliferation and, at the same time, for reducing a
capacity of the
hematopoietic mononuclear cells in responding to responding to signaling
pathways
involving PI-3 kinase, thereby expanding a population of the hematopoietic
stem
cells, while at the same time, substantially inhibiting differentiation of the
hematopoietic stem cells ex-vivo; and (c) transplanting or implanting the
hematopoietic stem cells to a recipient.
In yet another embodiment, the method comprises (a) obtaining hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells from a donor;
(b)
providing the hematopoietic mononuclear cells with ex-vivo culture conditions
for cell
proliferation and with nicotinamide, a nicotinamide analog, a nicotinamide or
a
nicotinamide analog derivative or a nicotinamide or a nicotinamide analog
metabolite, .
thereby expanding a population of the hematopoietic stem cells, while at the
same
time, substantially inhibiting differentiation of the hematopoietic stem cells
ex-vivo;
and (c) transplanting or implanting the hematopoietic stem cells to a
recipient.
In still yet another embodiment, the method comprises (a) obtaining
hematopoietic mononuclear cells which comprise a major fraction of
hematopoietic
committed cells and a minor fraction of hematopoietic stem and progenitor
cells from
a donor; (b) providing the hematopoietic mononuclear cells with ex-vivo
culture
conditions for cell proliferation and with a PI 3-kinase inhibitor, thereby
expanding a
population of the hematopoietic stem cells, while at the same time,
substantially
inhibiting differentiation of the hematopoietic stem cells ex-vivo; and (c)
transplanting
or implanting the hematopoietic stem cells to a recipient.
In yet another embodiment, the method comprises (a) obtaining hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells from a donor;
(b)



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
8
providing the hematopoietic mononuclear cells with ex-vivo culture conditions
for cell
proliferation and, at the same time, for reducing the expression and/or
activity of PI 3-
kinase, thereby expanding a population of the hematopoietic stem cells, while
at the
same time, substantially inhibiting differentiation of the hematopoietic stem
cells ex-
vivo; and (c) transplanting or implanting the hematopoietic stem cells to a
recipient.
In still yet another embodiment, the method comprises (a) obtaining
hematopoietic mononuclear cells which comprise a major fraction of
hematopoietic
committed cells and a minor fraction of hematopoietic stem and progenitor
cells from
a donor; (b) providing the hematopoietic mononuclear cells with ex-vivo
culture
conditions for cell proliferation and with one or more copper chelator(s) or
chelate(s),
thereby expanding a population of the hematopoietic stem cells, while at the
same
time, substantially inhibiting differentiation of the hematopoietic stem cells
ex-vivo;
and (c) transplanting or implanting the hematopoietic stem cells to a
recipient.
The donor and the recipient in the methods above can be a single individual or
different individuals, for example, allogeneic or xenogeneic individuals.
According to still another aspect of the present invention there are provided
transplantable hematopoietic cell preparations.
In one embodiment, a transplantable hematopoietic cell preparation of the
present invention comprises an expanded population of hematopoietic stem cells
propagated ex-vivo from hematopoietic mononuclear cells which comprise, prior
to
expansion, a major fraction of hematopoietic committed cells and a minor
fraction of
hematopoietic stem and progenitor cells, in the presence of an effective
amount of an
agent for reducing an expression and/or activity of CD38, while at the same
time,
substantially inhibiting differentiation of said hematopoietic stem cells, and
a
pharmaceutically acceptable carrier.
In another embodiment, a transplantable hematopoietic cell preparation of the
present invention comprises an expanded population of hematopoietic stem cells
propagated ex-vivo from hematopoietic mononuclear cells which comprise, prior
to
expansion, a major fraction of hematopoietic committed cells and a minor
fraction of
hematopoietic stem and progenitor cells, in the presence of an effective
amount of an
agent for reducing an expression and/or activity of PI 3-kinase, while at the
same
time, substantially inhibiting differentiation of said hematopoietic stem
cells, and a
pharmaceutically acceptable carrier.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
9
In still another embodiment, a transplantable hematopoietic cell preparation
of
the present invention comprises an expanded population of hematopoietic stem
cells
propagated ex-vivo from hematopoietic mononuclear cells which comprise, prior
to
expansion, a major fraction of hematopoietic committed cells and a minor
fraction of
hematopoietic stem and progenitor cells, in the presence of an effective
amount of an
agent, the agent reducing a capacity of the hematopoietic mononuclear cells in
responding to retinoic acid, retinoids and/or Vitamin D, while at the same
time,
substantially inhibiting differentiation of said hematopoietic stem cells, and
a
pharmaceutically acceptable carrier.
In yet another embodiment, a transplantable hematopoietic cell preparation of
the present invention comprises an expanded population of hematopoietic stem
cells
propagated ex-vivo from hematopoietic mononuclear cells which comprise, prior
to
expansion, a major fraction of hematopoietic committed cells and a minor
fraction of
hematopoietic stem and progenitor cells, in the presence of an effective
amount of an
agent, the agent reducing a capacity of the hematopoietic mononuclear cells in
responding to retinoic acid receptor, retinoid X receptor and/or Vitamin D
receptor
signaling, while at the same time, substantially inhibiting differentiation of
said
hematopoietic stem cells, and a pharmaceutically acceptable carrier.
In still another embodiment, a transplantable hematopoietic cell preparation
of
the present invention comprises an expanded population of hematopoietic stem
cells
propagated ex-vivo from hematopoietic mononuclear cells which comprise, prior
to
expansion, a major fraction of hematopoietic committed cells and a minor
fraction of
hematopoietic stem and progenitor cells, in the presence of an effective
amount of an
agent, the agent reducing a capacity of the hematopoietic mononuclear cells in
responding to PI 3-kinase signaling, while at the same time, substantially
inhibiting
differentiation of said hematopoietic stem cells, and a pharmaceutically
acceptable
carrier.
In yet another embodiment, a transplantable hematopoietic cell preparation of
the present invention comprises an expanded population of hematopoietic stem
cells
propagated ex-vivo from hematopoietic mononuclear cells which comprise, prior
to
expansion, a major fraction of hematopoietic committed cells and a minor
fraction of
hematopoietic stem and progenitor cells, in the presence of an effective
amount of an
agent selected from the group consisting of nicotinamide, a nicotinamide
analog, a



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
nicotinamide or a nicotinamide analog derivative and a nicotinamide or a
nicotinamide analog metabolite, while at the same time, substantially
inhibiting
differentiation of said hematopoietic stem cells, and a pharmaceutically
acceptable
carrier.
5 In still another embodiment, a transplantable hematopoietic cell preparation
of
the present invention comprises an expanded population of hematopoietic stem
cells
propagated ex-vivo from hematopoietic mononuclear cells which comprise, prior
to
expansion, a major fraction of hematopoietic committed cells and a minor
fraction of
hematopoietic stem and progenitor cells, in the presence of an effective
amount of a
10 PI 3-kinase inhibitor, while at the same time, substantially inhibiting
differentiation of
said hematopoietic stem cells, and a pharmaceutically acceptable carrier.
In yet another embodiment, a transplantable hematopoietic cell preparation of
the present invention comprises an expanded population of hematopoietic stem
cells
propagated ex-vivo from hematopoietic mononuclear cells which comprise, prior
to
expansion, a major fraction of hematopoietic committed cells and a minor
fraction of
hematopoietic stem and progenitor cells, in the presence of an effective
amount of one
or more copper chelator(s) or copper chelate(s), while at the same time,
substantially
inhibiting differentiation of said hematopoietic stem cells, and a
pharmaceutically
acceptable carrier.
According to an additional aspect of the present invention there is provided a
method of adoptive immunotherapy.
In one embodiment the method comprises (a) obtaining hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells from a
recipient; (b)
providing the hematopoietic mononuclear cells with ex-vivo culture conditions
for cell
proliferation and, at the same time, for reducing an expression and/or
activity of
CD38, thereby expanding a population of the hematopoietic stem cells, while at
the
same time, substantially inhibiting differentiation of the hematopoietic stem
cells; and
(c) transplanting said hematopoietic stem cells to the recipient.
3o In another embodiment the method comprises (a) obtaining hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells from a
recipient; (b)
providing the hematopoietic mononuclear cells with ex-vivo culture conditions
for cell



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
11
proliferation and, at the same time, for reducing a capacity of the
hematopoietic
mononuclear cells in responding to retinoic acid, retinoids and/or Vitamin D,
thereby
expanding a population of the stem cells, thereby expanding a population of
the
hematopoietic stem cells, while at the same time, substantially inhibiting
differentiation of the hematopoietic stem cells; and (c) transplanting said
hematopoietic stem cells to the recipient.
In still another embodiment the method comprises (a) obtaining hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells from a
recipient; (b)
l0 providing the hematopoietic mononuclear cells with ex-vivo culture
conditions for cell
proliferation and, at the same time, for reducing a capacity of the
hematopoietic
mononuclear cells in responding to signaling pathways involving the retinoic
acid
receptor and/or the retinoid X receptor and/or the Vitamin D receptor, thereby
expanding a population of the hematopoietic stem cells, while at the same
time,
substantially inhibiting differentiation of the hematopoietic stem cells; and
(c)
transplanting said hematopoietic stem cells to the recipient.
In yet another embodiment the method comprises (a) obtaining hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells from a
recipient; (b)
providing the hematopoietic mononuclear cells with ex-vivo culture conditions
for cell
proliferation and, at the same time, for reducing a capacity of the
hematopoietic
mononuclear cells in responding to signaling pathways involving PI 3-kinase,
thereby
expanding a population of the hematopoietic stem cells, while at the same
time,
substantially inhibiting differentiation of the hematopoietic stem cells; and
(c)
transplanting said hematopoietic stem cells to the recipient.
In still another embodiment the method comprises (a) obtaining hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells from a
recipient; (b)
providing the hematopoietic mononuclear cells with ex-vivo culture conditions
for cell
proliferation and with nicotinamide, a nicotinamide analog, a nicotinamide or
a
nicotinamide analog derivative or a nicotinamide or a nicotinamide analog
metabolite,
thereby expanding a population of the hematopoietic stem cells, while at the
same



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
12
time, substantially inhibiting differentiation of the hematopoietic stem
cells; and (c)
transplanting said hematopoietic stem cells to the recipient.
In yet another embodiment the method comprises (a) obtaining hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells from a
recipient; (b)
providing the hematopoietic mononuclear cells with ex-vivo culture conditions
for cell
proliferation and with a PI 3-kinase inhibitor, thereby expanding a population
of the
hematopoietic stem cells, while at the same time, substantially inhibiting
differentiation of the hematopoietic stem cells; and (c) transplanting said
hematopoietic stem cells to the recipient.
In still another embodiment the method comprises (a) obtaining hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells from a
recipient; (b)
providing the hematopoietic mononuclear cells with ex-vivo culture conditions
for cell
proliferation and with one or more copper chelator(s) or chelate(s), thereby
expanding
a population of the hematopoietic stem cells, while at the same time,
substantially
inhibiting differentiation of the hematopoietic stem cells; and (c)
transplanting said
hematopoietic stem cells to the recipient.
Further according to an aspect of the present invention, there is provided a
method of genetically modifying stem cells with an exogene.
In one embodiment, the method comprises (a) obtaining hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells; (b) providing
the
hematopoietic mononuclear cells with ex-vivo culture conditions for cell
proliferation
and, at the same time, for reducing an expression and/or activity of CD38,
thereby
expanding a population of the hematopoietic stem cells, while at the same
time,
substantially inhibiting differentiation of the hematopoietic stem cells ex-
vivo; and (c)
genetically modifying said hematopoietic stem cells with the exogene.
In another embodiment, the method comprises (a) obtaining hematopoietic
3o mononuclear cells which comprise a major fraction of hematopoietic
committed cells
and a minor fraction of hematopoietic stem and progenitor cells; (b) providing
the
hematopoietic mononuclear cells with ex-vivo culture conditions for cell
proliferation
and, at the same time, for reducing an expression and/or activity of PI 3-
kinase,



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
13
thereby expanding a population of the hematopoietic stem cells, while at the
same
time, substantially inhibiting differentiation of the hematopoietic stem cells
ex-vivo;
and (c) genetically modifying said hematopoietic stem cells with the exogene.
In still another embodiment, the method comprises (a) obtaining
hematopoietic mononuclear cells which comprise a major fraction of
hematopoietic
committed cells and a minor fraction of hematopoietic stem and progenitor
cells; (b)
providing the hematopoietic mononuclear cells with ex-vivo culture conditions
for cell
proliferation and, at the same time, for reducing a capacity of the
hematopoietic
mononuclear cells in responding to retinoic acid, retinoids and/or Vitamin D,
thereby
1 o expanding a population of the hematopoietic stem cells, while at the same
time,
substantially inhibiting differentiation of the hematopoietic stem cells ex-
vivo; and (c)
genetically modifying said hematopoietic stem cells with the exogene.
In yet another embodiment, the method comprises (a) obtaining hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells; (b) providing
the
hematopoietic mononuclear cells with ex-vivo culture conditions for cell
proliferation
and, at the same time, for reducing a capacity of the hematopoietic
mononuclear cells
in responding to signaling pathways involving the retinoic acid receptor
and/or the
retinoid X receptor and/or the Vitamin D receptor, thereby expanding a
population of
2o the hematopoietic stem cells, while at the same time, substantially
inhibiting
differentiation of the hematopoietic stem cells ex-vivo; and (c) genetically
modifying
said hematopoietic stem cells with the exogene.
In still another embodiment, the method comprises (a) obtaining
hematopoietic mononuclear cells which comprise a major fraction of
hematopoietic
committed cells and a minor fraction of hematopoietic stem and progenitor
cells; (b)
providing the hematopoietic mononuclear cells with ex-vivo culture conditions
for cell
proliferation and, at the same time, for reducing a capacity of the
hematopoietic
mononuclear cells in responding to signaling pathways involving PI 3-kinase,
thereby
expanding a population of the hematopoietic stem cells, while at the same
time,
3o substantially inhibiting differentiation of the hematopoietic stem cells ex-
vivo; and (c)
genetically modifying said hematopoietic stem cells with the exogene.
In yet another embodiment, the method comprises (a) obtaining hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
14
and a minor fraction of hematopoietic stem and progenitor cells; (b) providing
the
hematopoietic mononuclear cells with ex-vivo culture conditions for cell
proliferation
and with nicotinamide, a nicotinamide analog, a nicotinamide or a nicotinamide
analog derivative or a nicotinamide or a nicotinamide analog metabolite,
thereby
expanding a population of the hematopoietic stem cells, while at the same
time,
substantially inhibiting differentiation of the hematopoietic stem cells ex-
vivo; and (c)
genetically modifying said hematopoietic stem cells with the exogene.
In still another embodiment, the method comprises (a) obtaining
hematopoietic mononuclear cells which comprise a major fraction of
hematopoietic
committed cells and a minor fraction of hematopoietic stem and progenitor
cells; (b)
providing the hematopoietic mononuclear cells with ex-vivo culture conditions
for cell
proliferation and with a PI 3-kinase inhibitor, thereby expanding a population
of the
hematopoietic stem cells, while at the same time, substantially inhibiting
differentiation of the hematopoietic stem cells ex-vivo; and (c) genetically
modifying
said hematopoietic stem cells with the exogene.
In yet another embodiment, the method comprises (a) obtaining hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells; (b) providing
the
hematopoietic mononuclear cells with ex-vivo culture conditions for cell
proliferation
and with one or more copper chelator(s) or chelate(s), thereby expanding a
population
of the hematopoietic stem cells, while at the same time, substantially
inhibiting
differentiation of the hematopoietic stem cells ex-vivo; and (c) genetically
modifying
said hematopoietic stem cells with the exogene.
In a preferred embodiment, genetically modifying the cells is effected by a
vector which comprises the exogene, which vector is, for example, a viral
vector or a
nucleic acid vector.
According to still a further aspect of the present invention there is provided
a
hematopoietic stem cells collection/culturing bag supplemented with an
effective
amount of a retinoic acid receptor antagonist, a retinoid X receptor
antagonist and/or a
Vitamin D receptor antagonist, with an effective amount of nicotinamide, a
nicotinamide analog, a nicotinamide or a nicotinamide analog derivative and a
nicotinamide or a nicotinamide analog metabolite, with an effective amount of
a PI 3-
kinase inhibitor, or with an effective amount of a copper chelator or chelate,
each of



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
which substantially inhibits cell differentiation of a hematopoietic stem
cells fraction
of hematopoietic mononuclear cells which comprise a major fraction of
hematopoietic
committed cells and a minor fraction of hematopoietic stem and progenitor
cells.
According to an additional aspect of the present invention, there is provided
an
5 assay of determining whether an agent/molecule is an effective hematopoietic
stem
cell expansion agent. The assay comprises culturing hematopoietic mononuclear
cells
which comprise a major fraction of hematopoietic committed cells and a minor
fraction of hematopoietic stem and progenitor cells in the presence of tested
agent/molecule and monitoring expansion of the hematopoietic stem cells,
wherein if
10 increased expansion and decreased differentiation of the hematopoietic stem
cells
occurs, as compared to non-treated hematopoietic mononuclear cells, the tested
agent/molecule is an effective hematopoietic stem cell expansion agent.
The agent/molecule can be a retinoic acid receptor antagonist, a retinoid X
receptor antagonist, a Vitamin D receptor antagonist, nicotinamide and an
analog, a
15 derivative and a metabolite thereof, a PI 3-kinase inhibitor, a copper
chelator and a
copper chelate.
According to further features in preferred embodiments of the invention
described below, reducing the expression and/or activity of CD38 is effected
by an
agent that downregulates CD38 expression.
According to still further features in the described preferred embodiments the
agent that downregulates CD38 expression is selected from the group consisting
of a
retinoic acid receptor antagonist, a retinoid X receptor antagonist and a
Vitamin D
receptor antagonist. Alternatively, this agent is an antagonist for reducing a
capacity
of the stem cells in responding to retinoic acid, retinoid and/or Vitamin D.
Further
alternatively, the agent that downregulates CD38 expression is a PI 3-kinase
inhibitor.
According to still further features in the described preferred embodiments the
agent that downregulates CD38 expression is a polynucleotide.
According to still further features in the described preferred embodiments the
polynucleotide encodes an anti CD38, an anti retinoic acid receptor, an anti
retinoid X
receptor, an anti Vitamin D receptor or an anti PI 3-kinase antibody or
intracellular
antibody.
According to still further features in the described preferred embodiments the
polynucleotide is a small interfering polynucleotide molecule directed to
cause



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
16
intracellular CD38, retinoic acid receptor, retinoid X receptor, Vitamin D
receptor or
PI 3-kinase mRNA degradation.
According to still further features in the described preferred embodiments the
small interfering polynucleotide molecule is selected from the group
consisting of an
RNAi molecule, an anti-sense molecule, a rybozyme molecule and a DNAzyme
molecule.
According to further features in preferred embodiments of the invention
described below, reducing the expression and/or activity of CD38 is effected
by an
agent that inhibits CD38 activity. The agent can be, for example,
nicotinamide, a
nicotinamide analog, a nicotinamide or a nicotinamide analog derivative or a
nicotinamide or a nicotinamide analog metabolite. The nicotinamide analog is
preferably selected from the group consisting of benzamide, nicotinethioamide,
nicotinic acid and a,-amino-3-indolepropionic acid.
According to further features in preferred embodiments of the invention
described below, reducing the expression and/or activity of CD38 is effected
by an
agent that inhibits PI 3-kinase activity.
According to further features in preferred embodiments of the invention
described below, providing the stem cells with the conditions for ex-vivo cell
proliferation comprises providing the cells with nutrients and with cytokines.
According to still further features in the described preferred embodiments the
cytokines are early acting cytokines, such as, but not limited to, stem cell
factor, FLT3
ligand, interleukin-l, interleukin-2, interleukin-3, interleukin-6,
interleukin-10,
interleukin-12, tumor necrosis factor-a and thrombopoietin.
According to still further features in the described preferred embodiments the
cytokines are late acting cytokines, such as, but not limited to, granulocyte
colony
stimulating factor, granulocyte/macrophage colony stimulating factor,
erythropoietin,
FGF, EGF, NGF, VEGF, LIF, Hepatocyte growth factor and macrophage colony
stimulating factor.
According to still further features in the described preferred embodiments the
hematopoietic mononuclear cells are derived from a source selected from the
group
consisting of bone marrow, peripheral blood and neonatal umbilical cord blood.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
17
According to still further features in the described preferred embodiments
reducing the capacity of the hematopoietic mononuclear cells in responding to
signaling pathways is reversible, e.g., inherently reversible.
According to still further features in the described preferred embodiments
reducing the capacity of the hematopoietic mononuclear cells in responding to
the
above antagonists and/or signaling pathways of the above receptors is by ex-
vivo
culturing the hematopoietic mononuclear cells in a presence of an effective
amount of
at least one retinoic acid receptor antagonist, at least one retinoid X
receptor
antagonist and/or at least one Vitamin D receptor antagonist, preferably, for
a time
period of 0.1-50 %, preferably, 0.1-25 %, more preferably, 0.1-15 %, of an
entire ex-
vivo culturing period of the hematopoietic mononuclear cells.
According to still further features in the described preferred embodiments,
the
retinoic acid receptor antagonist is selected from the group consisting o~
AGN 194310; AGN 193109; 3-(4-Methoxy-phenylsulfanyl)-3-methyl-butyric acid;
6-Methoxy-2,2-dimethvl-thiochroman-4-one,2,2-Dimethyl-4-oxo-thiochroman-6-
yltrifluoromethane-sulfonate; Ethyl 4-((2,2 dimethyl-4-oxo-thiochroman-6-
yl)ethynyl)-benzoate; Ethyl 4-((2,2-dimethy 1-4-triflouromethanensulfonyloxy -
(2H)-
thiochromen-6-yl)ethynyl)-benzoate(41 ); Thiochromen-6-yl]-ethynyl]-
benzoate(yl);
(p-[(E)-2-[3'4'-Dihydro-4,4'-dimethyl-7'-(heptyloxy)-2'H-1-benzothiopyran-
6'yl]
propenyl] benzoic acid 1' 1'-dioxide; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
butoxyphenyl)-
3-methyl]-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
propoxyphenyl)-
3-methyl]-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
pentoxyphenyl)-
3-methyl]-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
hexoxyphenyl)-3-
methyl]-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
heptoxyphenyl)-3-
methyl]-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-
octoxyphenyl)-3-
methyl]-octa-2,4,6-trienoic acid; (2E,4E,6E)-7-[3-t-butyl-5-(1-phenyl-vinyl)-
phenyl]-
3-methyl-octa-2,4,6-trienoic acid; 2E,4E,6E-[7-(3,5-Di-t-butyl-4-{[4,5-3
H2]-n-pentoxy}phenyl)-3-methyl]-octa-2,4,6-trienoic acid; (2E,4E)-
(1RS,2RS)-
S-[2-(3,5-di-tert.butyl-2-ethoxy-phenyl)-cyclopropyl]-3-methyl-penta-2,4-
dienoic acid
ethyl ester; (2E,4E)-(1RS,2RS)-5-[2-(3,5-di-tert.butyl-2-ethoxy-phenyl)-
cyclopropyl]-
3-methyl-penta-2,4-dienoic acid; (2E,4E)-(1RS,2RS)-5-[2-(3,5-di-tert.butyl-2-
butoxy-phenyl)-cyclopropyl]-3-methyl-penta-2,4-dienoic acid; (2E,4E,6Z)-7-[3,5-
di-
tert.butyl-2-ethoxyphenyl]3-methyl-2,4,6-octatrienoic acid; (2E,4E,6Z)-7-[3,5-
di-



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
18
tert.butyl-2-butyloxyphenyl]-3-methyl-2,4,6-octatrienoic acid; 4-(5,6,7,8-
tetrahydro-
5,5,8,8-tetramethyl-2-naphthalene-carboxamido) benzoic acid; (2E,4E)-3-methyl-
5-
[( 1 S,2S)-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydro-naphthalen-2-yl)-
cyclopropyl]-
penta-2,4-dienoic acid; p-[(E)-2-[3',4'-Dihydro-4',4'-dimethyl-7'-(heptyloxy)-
2'H-1-
benzothiopyran-6'-yl]propenyl]benzoic acid; 1',1'-dioxide, 4-(7,7,10,10-
Tetramethyl-
1-pyridin-3-ylmethyl-4,5,7,8,9,10-hexahydro-1H-naphto[2,3-g]indol-3-yl)-
benzoic
acid; (2E,4E,6Z)-7-[3,5-di-tert.butyl-2-methoxyphenyl]-3-methyl-2,4,6-
octatrienoic
acid; (2E,4E,6Z)-7-[3,5-di-tert.butyl-2-ethoxyphenyl]-3-methyl-2,4,6-
octatrienoic
acid; (2E,4E,6Z)-7-[3,5-di-tert.butyl-2-hexyloxyphenyl]-3-methyl-2,4,6-
octatrienoic
to acid; (2E,4E,6Z)-7-[3,5-di-tert.butyl-2-octyloxyphenyl]-3-methyl-2,4,6-
octatrienoic
acid; and (2E,4E)-(1RS,2RS)-5-[2-(3,5-di-tert-butyl-2-butoxy-phenyl)-
cyclopropyl]-
3-methyl-penta-2,4-dienoic acid (2E,4E,6Z)-7-(3-n-propoxy-5,6,7,8-tetrahydro-
5,5,8,8-tetramethylnaphthalene-2-yl)-3-methylocta-2,4,6-trienoic acid, 4-(5H-
2,3(2,5
dimethyl-2,5-hexano)-5-n-propyldibenzo[b,e][1,4]diazepin-11-yl)benzoic acid, 4-

(5H-2,3-(2,5-dimethyl-2,5-hexano)-5methyl-8-nitrodibenzo[b,e][1,4]diazepin-11-
yl)benzoic acid, 4-{[4-(4-Ethylphenyl)2,2-dimethyl-(2H)-thiochromen-6-
yl]ethynyl}benzoic acid, 4-[4-2methyl-1,2-dicarba-closo-dodecaboran-1-yl-
phenylcarbamoyl]benzoic acid, 4-[4,5,7,8,9,10-hexahydro-7,7,10,10-tetramethyl-
1-(3-
pyridylmethyl)-anthra[1,2-b]pyrrol-3-yl]benzoic acid, (3-pyridylmethyl)-]5-
thiaanthra[2,1-b]pyrrol-3-yl)benzoic acid, and (3-pyridylmethyl)-anthra[2m1-
d]pyrazol-3-yl]benzoic acid.
According to still further features in the described preferred embodiments,
the
retinoid X receptor antagonist is selected from the group consisting of:
LGN100572, LGN100574, 1-(3-hydroxy-5,6,7,8-tetrahydro-5,5,8,8-
tetramethylnaphthalene-2-yl)ethanone, 1-(3-propoxy-5,6,7,8-tetrahydro-5,5,8,8-
tetramethylnaphthalene-2-yl)ethanone, 3-(3-propoxy-5,6,7,8-tetrahydro-5,5,8,8-
tetramethylnaphthalene-2-yl)but-2-enenitrile, 3-(3-propoxy-5,6,7,8-tetrahydro-
5,5,8,8-tetramethylnaphthalene-2-yl)but-2-enal, (2E,4E,6E)-7-3[-propoxy-
5,6,7,8-
tetrahydro 5,5,8,8-tetramethyl-2-naphthalene-2-yl]-3-methylocta-2,4,6-trienoic
acid,
4-[3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)carbonyl] benzoic acid,
4-[1-
(3,5, 5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)ethenyl] benzoic acid, 4-

[1(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)cyclopropyl] benzoic
acid, 4-
[1-(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)ethenyl] benzenete
trazole,



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
19
2-[1-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydro-2-naphthyl) ethenyl]pyridine-5-
carboxylic acid, 2-[1-(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-
naphthyl)ethyl]pyridine-5-carboxylic acid, ethyl-2-[1-(3,5,5,8, 8-pentamethyl-
5,6,7,8-
tetrahydro-2-naphthyl)ethenyl]pyridine-5-carboxylate, 5-[1-3,5,5,8,8-
pentamethyl-
5,6,7,8-tetrahydro-2-naphthyl)ethenyl]pyridine-2-carboxylic acid, 2-[1-
(3,5,5,8,8-
pentamethyl-5,6,7,8-tetrahydro-2-naphthyl) cyclopropyl]pyridine-5-carboxylic
acid,
methyl 2-[1-(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-
naphthyl)cyclopropyl]pyridine-5-carboxylate, 4-[1-(3,5, 5,8,8-pentamethyl-
5,6,7,8-
tetrahydro-2-naphthyl)ethenyl]-N-(4-hydroxyphenyl) benzamide, 2-[1-(3,5,5,8,8-
Pentamethyl-5,6,7,8-tetrahydro-2-naphthyl) ethenyl] pyridine-5-carboxylic
acid, 2-[1-
(3,5,5,8,8-Pentamethyl-5, 6,7,8-tetrahydro-2-naphthyl)cyclopropyl]pyridine-5-
carboxylic acid, 4-[(3,5, 5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-
naphthyl)carbonyl]benzoic acid butyloxime, 4-[(3,5,5,8,8-pentamethyl-5,6,7,8-
tetrahydro-2-naphthyl) carbonyl]benzoic acid propyloxime, 4-[(3,5,5,8,8-
pentamethyl-5,6,7,8-terrahydro-2-naphthyl)carbonyl]benzoic acid cyanoimine, 4-
[(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)carbonyl]benzoic acid
allyloxime, 4-[(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-
naphthyl)carbonyl]benzoic
acid 4-(3-methylbut-2-enoic acid)oxime, and 4-[(3,5,5,8,8-pentamethyl-5,6,7,8-
tetrahydro-2-naphthyl)carbonyl]benzoic acid 1-aminoethyloxime (2E,4E,6Z)-7-(3-
n-
propoxy-5,6,7,8-tetrahydro-5,5,8,8-tetramethylnaphthalene-2-yl)-3-methylocta-
2,4,6-
trienoic acid, and 4-(5H-2,3(2,5 dimethyl-2,5-hexano)-5-n-
propyldibenzo[b,e][1,4]diazepin-11-yl)benzoic acid, and 4-(5H-2,3-(2,5-
dimethyl-
2,5-hexano)-5methyl-8-nitrodibenzo[b,e][1,4]diazepin-11-yl)benzoic acid.
According to still further features in the described preferred embodiments,
the
Vitamin D receptor antagonist is selected from the group consisting of: 1
alpha, 25-
(OH)-D3-26,23 lactone; lalpha, 25-dihydroxyvitamin D (3); the 25-carboxylic
ester
ZK159222; (23S)- 25-dehydro-1 alpha-OH-D (3); (23R)-25-dehydro-1 alpha-OH-D
(3); 1 beta, 25 (OH)2 D3; 1 beta, 25(OH)Z-3-epi-D3; (23S) 25-dehydro-1
alpha(OH)
D3-26,23-lactone; (23R) 25-dehydro-1 alpha(OH)D3-26,23-lactone and Butyl-
(5Z,7E,22E-( 1 S,7E,22E-( 1 S,3R,24R)-1,3,24-trihydroxy-26,27-cyclo-9,10-
secocholesta-5,7,10( 19),22-tetraene-25-carboxylate).



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
According to still further features in the described preferred embodiments,
the
PI 3-kinase inhibitor is selected from the group consisting of wortmannin and
LY294002.
The copper chelate(s) or chelator(s) used in the various aspects of the
present
5 invention described hereinabove preferably comprise a polyamine chelator.
According to further features in preferred embodiments of the invention
described below, the polyamine chelator is capable of forming an
organometallic
complex with a transition metal other than copper. The transition metal can
be, for
example, zinc, cobalt, nickel, iron, palladium, platinum, rhodium and
ruthenium.
1 o According to still further features in the described preferred embodiments
the
polyamine chelator is a linear polyamine.
Preferably, the linear polyamine has a general formula I:
HX-Am-(Y~B~)~ ~~~~(YnBn)n-ZH
15 Formula I
wherein m is an integer from 1 to 10; n is an integer from 0 to 20; X and Z
are each
independently selected from the group consisting of an oxygen atom, a sulfur
atom
and a -NH group; Y, and Yn are each independently selected from the group
20 consisting of an oxygen atom, a sulfur atom and a -NH group; A is an
alkylene chain
having between 1 and 10 substituted and/or non-substituted carbon atoms; and
B, and
Bn are each independently an alkylene chain having between 1 and 20
substituted
and/or non-substituted carbon atoms, provided that at least one of the X, Z,
Y, and Yn
is a -NH group and/or at least one of the carbon atoms in the alkylene chains
is
substituted by an amine group.
According to still further features in the described preferred embodiments, A
is an alkylene chain having a general formula II:



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
21
Ri Rz Rg
-C~H-CzH.......CgH_
Formula II
wherein g is an integer that equals 0 or 3-10; and each of R~, RZ and Rg is
independently selected from the group consisting of hydrogen, alkyl, alkenyl,
alkynyl,
aryl, cycloalkyl, heteroalicyclic, heteroaryl, halo, amino, alkylamino,
arylamino,
cycloalkylamino, heteroalicyclic amino, heteroarylamino, hydroxy, alkoxy,
aryloxy,
azo, C-amido, N-amido, ammonium, thiohydroxy, thioalkoxy, thioaryloxy,
sulfonyl,
sulfinyl, N-sulfonamide, S-sulfonamide, phosphonyl, phosphinyl, phosphonium,
carbonyl, thiocarbonyl, C-carboxy, O-carboxy, C-thiocarboxy, O-thiocarboxy, N-
carbamate, O-carbamate, N-thiocarbamate, O-thiocarbamate, urea, thiourea,
borate,
borane, boroaza, silyl, siloxy, silaza, aquo, alcohol, peroxo, amine oxide,
hydrazine,
alkyl hydrazine, aryl hydrazine, nitric oxide, cyanate, thiocyanate,
isocyanate,
isothiocyanate, cyano, alkylnitrile, aryl nitrile, alkyl isonitrile, aryl
isonitrile, nitrate,
nitrite, azido, alkyl sulfonic acid, aryl sulfonic acid, alkyl sulfoxide, aryl
sulfoxide,
alkyl aryl sulfoxide, alkyl sulfenic acid, aryl sulfenic acid, alkyl sulfuric
acid, aryl
sulfinic acid, alkyl thiol carboxylic acid, aryl thiol carboxylic acid, alkyl
thiol
thiocarboxylic acid, aryl thiol thiocarboxylic acid, carboxylic acid, alkyl
carboxylic
acid, aryl carboxylic acid, sulfate, sulfite, bisulfate, thiosulfate,
thiosulfite, alkyl
phosphine, aryl phosphine, alkyl phosphine oxide, aryl phosphine oxide, alkyl
aryl
phosphine oxide, alkyl phosphine sulfide, aryl phosphine sulfide, alkyl aryl
phosphine
sulfide, alkyl phosphonic acid, aryl phosphonic acid, alkyl phosphinic acid,
aryl
phosphinic acid, phosphate, thiophosphate, phosphate, pyrophosphate,
triphosphate,
hydrogen phosphate, dihydrogen phosphate, guanidino, S-dithiocarbamate, N-
dithiocarbamate, bicarbonate, carbonate, perchlorate, chlorate, chlorite,
hypochlorite,
perbromate, bromate, bromite, hypobromite, tetrahalomanganate,
tetrafluoroborate,
hexafluoroantimonate, hypophosphite, iodate, periodate, metaborate,
tetraarylborate,
tetraalkyl borate, tartarate, salicylate, succinate, citrate, ascorbate,
saccharirate, amino
acid, hydroxamic acid and thiotosylate.
According to still further features in the described preferred embodiments,
each of B 1 and Bn is independently an alkylene chain having a general formula
III:



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
22
Rp R~p+ 1 ) R9
-Cp-C(p+1)H .......CqH_
H
Formula III
wherein p is an integer that equals 0 or g+1; q is an integer from g+2 to
g+20; and
each of Rp, Rp+1 and Rq is independently selected from the group consisting of
the
substituents described hereinabove with respect to R,, RZ and Rg.
According to still further features in the described preferred embodiments at
least one of C~, CZ and Cg and/or at least one of Cp, Cp+1 and Cq is a chiral
carbon
atom.
A preferred linear polyamine according to the present invention is
tetraethylenepentamine.
According to still further features in the described preferred embodiments the
polyamine chelator is a cyclic polyamine, such as cyclam.
According to still further features in the described preferred embodiments the
cyclic polyamine has a general formula IV:
X Am-(Y~Bi)~---(YnBn)n-Z
Formula IV
wherein m is an integer from 1 to 10; n is an integer from 0 to 20; X and Z
are each
independently selected from the group consisting of an oxygen atom, a sulfur
atom
and a -NH group; Y~ and Yn are each independently selected from the group
consisting of an oxygen atom, a sulfur atom and a -NH group; A is an alkylene
chain
having between 1 and 10 substituted and/or non-substituted carbon atoms; B ~
and Bn
are each independently an alkylene chain having between 1 and 20 substituted
and/or
non-substituted carbon atoms; and D is a bridging group having a general
formula V:
U-W-V
Formula V
whereas U and V are each independently selected from the group consisting of
substituted hydrocarbon chain and non-substituted hydrocarbon chain; and W is
selected from the group consisting of amide, ether, ester, disulfide,
thioether,
thioester, imine and alkene,



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
23
provided that at least one of the X, Z, Y, and Yn is a -NH group and/or at
least
one of the carbon atoms in the alkylene chains is substituted by an amine
group.
According to still further features in the described preferred embodiments, A
and each of B 1 and Bn in Formula IV are alkylene chains having the general
formulas
II and III, as is described hereinabove.
According to still further features in the described preferred embodiments the
cyclic polyamine has a general formula selected from the group consisting of:
X Am-(Y~B~)~---(YnBn)n-ZH
Formula VI
D
HX-Am-(Y ~ B 1 ) ~- - - (YnBn)n-Z
Formula VII
______
X Am-(Y~B~)~---(YnBn)n-ZH
Formula VIII
______
HX-Am-(Y ~ B ~ ) ~- _ _ (Yn Bn)n-Z
Formula IX
.D____T________
HX-Am-(YIBI)1---(YnBn)n-ZH
Formula X
wherein m is an integer from 1 to 10; n is an integer from 0 to 20; X and Z
are each
independently selected from the group consisting of an oxygen atom, a sulfur
atom
and a -NH group; Y, and Yn are each independently selected from the group
consisting of an oxygen atom, a sulfur atom and a -NH group; A is an alkylene
chain
having between 1 and 10 substihited and/or non-substituted carbon atoms; B 1
and Bn



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
24
are each independently an alkylene chain having between 1 and 20 substituted
and/or
non-substituted carbon atoms; and D is a bridging group having a general
formula V,
as described hereinabove, and further wherein should the D is attached at one
end to
A (Formulas VI, VII and X), the U or the V are being attached to one carbon
atom in
the alkylene chain and should the D is attached at one end to B 1 or Bn
(Formulas
VIII, IX and X), the U or the V are being attached to one carbon atom in the
alkylene
chain,
provided that at least one of the X, Z, Y, and Yn is a -NH group and/or at
least
one of the carbon atoms in the alkylene chains is substituted by an amine
group.
1o The alkylene chains A, B1 and Bn are preferably as described hereinabove.
According to still further features in the described preferred embodiments the
polyamine chelator includes at least one linear polyamine and at least one
cyclic
polyamine.
Such a polyamine chelator preferably has a general formula XI:
f(EI)rfQl-(GI)g~~h-i(E2)i LQ2'(G2)J~~k_......._~(En)IyQn-(Gn)o~~t
Formula XI
wherein n is an integer greater than 1; each of f, g, h, i, j, k, l, o and t
is independently
an integer from 0 to 10; each of EI, EZ and En is independently a linear
polyamine as
is described hereinabove; each of GI, GZ and Gn is independently a cyclic
polyamine
as is described hereinabove; and each of QI, QZ and Qn is independently a
linker
linking between two of the polyamines,
provided that at least one of the Q,, QZ and Qn is an amine group and/or at
least one of the linear polyamine and the cyclic polyamine is having at least
one free
amore group.
According to still further features in the described preferred embodiments
each
of Q,, QZ and Qn is independently selected from the group consisting alkylene,
alkenylene, alkynylene, arylene, cycloalkylene, hetroarylene, amine, azo,
amide,
sulfonyl, sulfinyl, sulfonamide, phosphonyl, phosphinyl, phosphonium,
ketoester,
carbonyl, thiocarbonyl, ester, ether, thioether, carbamate, thiocarbamate,
urea,
thiourea, borate, borane, boroaza, silyl, siloxy and silaza.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
According to still further features in the described preferred embodiments the
polyamine chelator is selected from the group consisting of ethylendiamine,
diethylenetriamine, triethylenetetramine, triethylenediamine,
tetraethylenepentamine,
aminoethylethanolamine, aminoethylpiperazine, pentaethylenehexamine,
captopril,
5 penicilamine, N,N'-bis(3-aminopropyl)-1,3-propanediamine, N,N'-Bis-(2-
animoethyl)-1,3-propanediamine, 1,7-dioxa-4,10-diazacyclododecane, 1,4,8,11-
tetraaza cyclotetradecane-5,7-dione, 1,4,7-triazacyclononane, 1-oxa-4,7,10-
triazacyclododecane, 1,4,8,12-tetraazacyclopentadecane, and 1,4,7,10-
tetraazacyclododecane.
10 The present invention successfully addresses the shortcomings of the
presently
known configurations by providing methods of expanding hematopoietic stem
cells
without first enriching hematopoietic mononuclear cells for stem cells.
Unless otherwise defined, all technical and scientific terms used herein have
the same meaning as commonly understood by one of ordinary skill in the art to
which
15 this invention belongs. Although methods and materials similar or
equivalent to those
described herein can be used in the practice or testing of the present
invention, suitable
methods and materials are described below. In case of conflict, the patent
specification, including definitions, will control. In addition, the
materials, methods,
and examples are illustrative only and not intended to be limiting.
BRIEF DESCRIPTION OF THE DRAWINGS
The invention is herein described, by way of example only, with reference to
the accompanying drawings. With specific reference now to the drawings in
detail, it
is stressed that the particulars shown are by way of example and for purposes
of
illustrative discussion of the preferred embodiments of the present invention
only, and
are presented in the cause of providing what is believed to be the most useful
and
readily understood description of the principles and conceptual aspects of the
invention. In this regard, no attempt is made to show structural details of
the invention
in more detail than is necessary for a fundamental understanding of the
invention, the
description taken with the drawings making apparent to those skilled in the
art how the
several forms of the invention may be embodied in practice.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
In the drawings:
26
FIGS. la-b illustrates the effect of TEPA chelator on the expansion of CD34+
hematopoietic stem cells in a culture of hematopoietic mononuclear cells. Cord-
blood
mononuclear cells (MNCs) were seeded in culture-bags in the presence of
cytokines,
and were either supplemented with TEPA chelator (MNC-TEPA), or not
supplemented with TEPA chelator (MNC control). For comparison, purified CD34+
cells were similarly seeded in culture-bags in the presence of cytokines with
no
supplementation of TEPA chelator (CD34+ culture). All cultures were incubated
for
12 weeks and at weekly intervals, the CD34+ cells were purified from cultures
using
miniMacs columns and enumerated;
FIG. 2 illustrates the FACS-analysis of the density of CD34+CD38- cells in the
untreated NMCs, TEPA-treated MNCs and CD34+ cell cultures described above; and
FIG. 3 presents the comparative numbers of colony-forming cells (CFUs)
measured from the untreated MNCs, TEPA-treated MNCs and CD34+ cell cultures
described above, at weekly intervals.
DESCRIPTION OF THE PREFERRED EMBODIMENTS
The present invention is of methods of ex-vivo expanding a population of
hematopoietic stem cells present, as a minor fraction, in hematopoietic
mononuclear
cells, without first enriching the stem cells, while at the same time,
substantially
inhibiting differentiation of the hematopoietic stem cells. The present
invention can
be used to efficiently provide ex-vivo expanded populations of hematopoietic
stem
cells, using hematopoietic mononuclear cells that comprise a major fraction of
hematopoietic committed cells and a minor fraction of the hematopoietic stem
and
progenitor cells as a source of stem cells, without prior enrichment of the
hematopoietic mononuclear cells for stem cells. The expanded populations of
hematopoietic stem cells of the present invention can be used in, for example,
hematopoietic cell transplantation, in generation of stem cells suitable for
genetic
manipulations for cellular gene therapy, as well as in additional application
such as,
but not limited to, adoptive immunotherapy, implantation of stem cells in an
in vivo
cis-differentiation and trans-differentiation settings, as well as, ex-vivo
tissue
engineering in cis-differentiation and trans-differentiation settings.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
27
The methods of the present invention utilize various molecules (also referred
to herein as agents), that interfere with CD38 expression and/or activity
and/or with
intracellular copper content, for inducing the ex-vivo expansion of
hematopoietic stem
cell populations described above, thereby providing an efficient, simplified
and yet
versatile technology for ex-vivo expansion of hematopoietic stem cells.
The principles and operation of the present invention may be better understood
with reference to the drawings and accompanying descriptions and examples.
Before explaining at least one embodiment of the invention in detail, it is to
be
understood that the invention is not limited in its application to the details
of
construction and the arrangement of the components set forth in the following
description or illustrated in the Examples section. The invention is capable
of other
embodiments or of being practiced or carried out in various ways. Also, it is
to be
understood that the phraseology and terminology employed herein is for the
purpose
of description and should not be regarded as limiting.
1s As is discussed hereinabove, WO 99/40783, WO 00/18885 and Peled et al,
Brit. J. Haematol. 116:655 2002 teach that cellular copper is involved in
modulating
the balance between self renewal and differentiation of hematopoietic
progenitor
cells. According to the teachings of these references, the addition of
transition metal
chelators that are capable of binding copper, such as, for example, the linear
polyamine tetraethylenepentamine, to CD34+ cell cultures in the presence of
early
acting cytokines reduced cell copper content by 30 % and extended the duration
of the
long-term cultures in terms of long-term CFU and CD34+ cell expansion. These
references hence teach methods of expanding stem cell populations, ex-vivo in
the
presence of transition metal chelators, copper chelators in particular, and
further teach
2s the use of the obtained expanded stem cell populations in various
applications.
PCT/IL03/00062 discloses that copper chelates, namely, copper chelators that
are complexed with a copper ion, also promote proliferation and inhibit
differentiation
of stem and progenitor cells when added to the culture media of such cells.
According to the teachings of PCT/IL03/00062, these finding suggest that this
effect
of copper chelates on proliferation and differentiation of stem and progenitor
cells is
not associated solely with the content of cellular copper but rather with
additional
regulatory pathways.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
28
PCT/IL03/00064 and U.S. Provisional Patent Application No. 60/452,545,
which are incorporated by reference as if fully set forth herein, disclose
that a series of
molecules that are capable of interfering with CD38 expression and/or
activity,
repress the process of differentiation of stem cells and stimulates and
prolongs, for up
to 16-18 weeks, the phase of active cell proliferation and expansion (renewal)
ex-vivo,
in a reversible manner. Hence, these references teach methods of expanding
stem cell
populations ex-vivo, which involve the addition of agents that either
downregulate
CD38 expression or inhibit the activity of CD38 to the culture media of stem
cells.
The methods disclosed in PCT/IL03/00064 and U.S. Provisional Patent
Application
No. 60/452,545, therefore utilize molecules such as retinoic acid receptor
antagonists
of the RAR and RXR superfamilies, Vitamin D receptor antagonists,
polynucleotides
encoding antibodies such as anti CD38, anti retinoic acid receptor, anti
retinoid X
receptor, anti Vitamin D receptor, polynucleotides that are directed to cause
degradation of endogenous polynucleotides encoding for these receptors,
molecules
that are capable of interfering with expression and/or activity of PI 3-kinase
and
CD38 inhibitors such as nicotinamide and its related compounds.
Hence, WO 99/40783, WO 00/18885, PCT/IL03/00064 and U.S. Provisional
Patent Application No. 60/452,545 all teach the use of various molecules that
modulate, via diverse pathways and/or mechanisms, the balance between self
renewal
and differentiation of stem cells, hematopoietic stem cells in particular, in
methods for
ex-vivo expanding of stem cell populations. However, unless otherwise
indicated, the
regulation of self renewal and differentiation of stem cells by these
molecules is
obtained, according to the teachings of these references, when the cultured
cells are
first enriched for stem and/or progenitor cells and hence, in line with other
present
day technologies in this field, require preliminary stem cells enrichment.
While reducing the present invention to practice, it was surprisingly and
unexpectedly found that molecules such as copper chelators, copper chelates
and
retinoic acid receptor (RAR) antagonists repress differentiation and stimulate
and
prolong proliferation of hematopoietic stem cells when the source of cells
includes the
3o entire fraction of mononuclear blood cells, namely non-enriched stem cells.
As is described in the Background section hereinabove, although being highly
advantageous, presently there is no disclosed technology by which to expand
non-
enriched stem cells. Therefore, the technology presented and exemplified
herein,



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
29
involving methods of ex-vivo expanding hematopoietic stem cell populations
devoid
of prior stem cells enrichment, provides for efficient, simplified and cost-
effective
methods of obtaining ex-vivo expanded hematopoietic stem cell populations. The
expanded hematopoietic stem cell populations obtained by the technology
presented
herein can be used in various application, the following lists a few:
Hematopoietic cell transplantation: Transplantation of hematopoietic cells has
become the treatment of choice for a variety of inherited or malignant
diseases. While
early transplantation procedures utilized the entire bone marrow (BM)
population,
recently, more defined populations, enriched for stem cells (CD34+ cells) have
been
used (Van Epps DE, et al. Harvesting, characterization, and culture of CD34+
cells
from human bone marrow, peripheral blood, and cord blood. Blood Cells 20:411,
1994). In addition to the marrow, such cells could be derived from other
sources such
as peripheral blood (PB) and neonatal umbilical cord blood (CB) (Emerson SG.
Ex-
vivo expansion of hematopoietic precursors, progenitors, and stem cells: The
next
~ 5 generation of cellular therapeutics. Blood 87:3082, 1996). Compared to BM,
transplantation with PB cells shortens the period of pancytopenia and reduces
the risks
of infection and bleeding (Brugger W, et al. Reconstitution of hematopoiesis
after
high-dose chematotherapy by autologous progenitor cells generated in-vivo. N
Engl J
Med 333:283, 1995; Williams SF, et al. Selection and expansion of peripheral
blood
CD34+ cells in autologous stem cell transplantation for breast cancer. Blood
87:1687,
1996; Zimmerman RM, et al. Large-scale selection of CD34+ peripheral blood
progenitors and expansion of neutrophil precursors for clinical applications.
J
Heamatotherapy 5:247, 1996).
An additional advantage of using PB for transplantation is its accessibility.
The limiting factor for PB transplantation is the low number of circulating
pluripotent
stem/progenitor cells.
To obtain enough PB-derived stem cells for transplantation, these cells are
"harvested" by repeated leukophoresis following their mobilization from the
marrow
into the circulation by treatment with chemotherapy and cytokines (Brugger W,
et al.
Reconstitution of hematopoiesis after high-dose chematotherapy by autologous
progenitor cells generated in-vivo. N Engl J Med 333:283, 1995; Williams SF,
et al.
Selection and expansion of peripheral blood CD34+ cells in autologous stem
cell



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
transplantation for breast cancer. Blood 87:1687, 1996). Such treatment is
obviously
not suitable for normal donors.
The use of ex-vivo expanded stem cells for transplantation has the following
advantages (Koller MR, Emerson SG, Palsson BO. Large-scale expansion of human
5 stem and progenitor cells from bone marrow mononuclear cells in continuous
perfusion cultures. Blood 82:378, 1993; Lebkowski JS, et al. Rapid isolation
and
serum-free expansion of human CD34+ cells. Blood Cells 20:404, 1994):
It reduces the volume of blood required for reconstitution of an adult
hematopoietic system and may obviate the need for mobilization and
leukophoresis
10 (Brugger W, et al. N Engl J Med 333:283, 1995).
It enables storage of small number of PB or CB stem cells for potential future
use.
In the case of autologous transplantation of recipients with malignancies,
contaminating tumor cells in autologous infusion often contribute to the
recurrence of
15 the disease (Brugger W, et al. N Engl J Med 333:283, 1995). Selecting and
expanding CD34+ stem cells will reduce the load of tumor cells in the final
transplant.
The cultures provide a significant depletion of T lymphocytes, which may be
useful in the allogeneic transplant setting for reducing graft-versus-host
disease.
Clinical studies indicate that transplantation of ex-vivo expanded cells
derived
20 from a small number of PB CD34+ cells can restore hematopoiesis in
recipients
treated with high doses of chemotherapy, although the results do not yet allow
firm
conclusions about long term in-vivo hematopoietic capabilities of these
cultured cells
(Brugger W, et al. N Engl J Med 333:283, 1995; Williams SF, et al. Blood
87:1687,
1996).
25 For successful transplantation, shortening of the duration of the cytopenic
phase, as well as long-term engraftment, is crucial. Inclusion of intermediate
and late
progenitor cells in the transplant could accelerate the production of donor-
derived
mature cells thereby shortening the cytopenic phase. It is important,
therefore, that ex-
vivo expanded cells include, in addition to stem cells, more differentiated
progenitor
30 cells in order to optimize short-term recovery and long-term restoration of
hematopoiesis. Expansion of intermediate and late progenitor cells, especially
those
committed to the neutrophilic and megakaryocytic lineages, concomitant with
expansion of stem cells, should serve this purpose (Sandstrom CE, et al.
Effects of



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
31
CD34+ cell selection and perfusion on ex-vivo expansion of peripheral blood
mononuclear cells. Blood 86:958, 1995).
Such cultures may be useful in restoring hematopoiesis in recipients with
completely ablated bone marrow, as well as in providing a supportive measure
for
shortening recipient bone marrow recovery following conventional radio- or
chemotherapies.
Prenatal diagnosis of genetic defects in scarce cells: Prenatal diagnosis
involves the collection of embryonic cells from a pregnant woman, in utero,
and
analysis thereof for genetic defects. A preferred, non-invasive, means of
collecting
to embryonic cells involves separation of embryonic nucleated red blood cell
precursors
that have infiltrated into peripheral maternal circulation. However, since the
quantities of these cells are quite scarce, a further application of the
present invention
would be the expansion of such cells according to methods described herein,
prior to
analysis. The present invention, therefore, offers a means to expand embryonic
cells
for applications in prenatal diagnosis.
Gene therapy: For successful long-term gene therapy, a high frequency of
genetically modified stem cells with transgenes stably integrated within their
genome,
is an obligatory requirement. In BM tissue, while the majority of cells are
cycling
progenitors and precursors, stem cells constitute only a small fraction of the
cell
population and most of them are in a quiescent, non-cycling state. Viral-based
(e.g.,
retroviral) vectors require active cell division for integration of the
transgene into the
host genome. Therefore, gene transfer into fresh BM stem cells is highly
inefficient.
The ability to expand a purified population of stem cells and to regulate
their cell
division ex-vivo would provide for an increased probability of their genetic
modification (Palmiter RD. Regulation of metallothionein genes by heavy metals
appears to be mediated by a zinc-sensitive inhibitor that interacts with a
constitutively
active transcription factor, MTF-1. Proc Natl Acad Sci USA 91(4): 1219-1223,
1994).
Adoptive immunotherapy: Ex-vivo-expanded, defined lymphoid
subpopulations have been studied and used for adoptive immunotherapy of
various
malignancies, immunodeficiencies, viral and genetic diseases (Freedman AR, et
al.
Generation of T lymphocytes from bone marrow CD34+ cells in-vitro. Nature
Medicine 2: 46, 1996; Heslop HE, et al. Long term restoration of immunity
against



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
32
Epstein-Barr virus infection by adoptive transfer of gene-modified virus-
specific T
lymphocytes. Nature Medicine 2: 551, 1996; Protti MP, et al. Particulate
naturally
processed peptides prime a cytotoxic response against human melanoma in-vitro.
Cancer Res 56: 1210, 1996).
The treatment enhances the required immune response or replaces deficient
functions. This approach was pioneered clinically by Rosenberg et al.
(Rosenberg SA,
et al. Prospective randomized trial of high-dose interleukin-2 alone or in
conjunction
with lymphokine-activated killer cells for the treatment of patients with
advanced
cancer. J Natl Cancer Inst 85: 622, 1993) using a large number of autologous
ex-vivo
expanded non-specific killer T cells, and subsequently ex-vivo expanded
specific
tumor infiltrating lymphocytes.
Functionally active, antigen-presenting cells could be grown from a starting
population of CD34+ PB cells in cytokine-supported cultures, as well. These
cells can
present soluble protein antigens to autologous T cells in-vitro and, thus,
offer new
prospects for the immunotherapy of minimal residual disease after high dose
chemotherapy. Ex-vivo expansion of antigen-presenting dendritic cells has been
studied as well, and is an additional promising application of the currently
proposed
technology (Bernhard H, et al. Generation of immunostimulatory dendritic cells
from
human CD34+ hematopoietic progenitor cells of the bone marrow and peripheral
blood. Cancer Res 10: 99, 1995; Fisch P, et al. Generation of antigen-
presenting cells
for soluble protein antigens ex-vivo from peripheral blood CD34+ hematopoietic
progenitor cells in cancer patients. Eur J Immunol 26: 595, 1996; Siena S, et
al.
Massive ex-vivo generation of functional dendritic cells from mobilized CD34+
blood
progenitors for anticancer therapy. Expt Hematol 23:1463, 1996).
As is discussed in brief hereinabove and is further detailed in WO 99/40783,
WO 00/18885, PCT/IL03/00064 and U.S. Provisional Patent Application No.
60/452,545, copper chelators, copper chelates and retinoid receptor
antagonists, each
modulate the self renewal of stem cells via a different pathway, effecting
different
cellular events that lead to reduced differentiation and extended
proliferation of stem
cells. These molecules therefore represent a wide variety of molecules that
are
capable of inducing the effect of expanding a hematopoietic stem cells
population that
is present in a mixed hematopoietic cells population.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
33
Hence, according to one aspect of the present invention there is provided a
method of ex-vivo expanding a population of hematopoietic stem cells, while at
the
same time, substantially inhibiting differentiation of the stem cells ex-vivo.
The
method according to this aspect of the present invention is effected by
providing
hematopoietic mononuclear cells which comprise a major fraction of
hematopoietic
committed cells and a minor fraction of hematopoietic stem and progenitor
cells, with
ex-vivo culture conditions for ex-vivo cell proliferation and, at the same
time, for
reducing an expression and/or activity of CD38, thereby expanding a population
of
hematopoietic stem cells, while at the same time, substantially inhibiting
differentiation of the hematopoietic stem cells ex-vivo.
As used herein, the phrase "hematopoietic mononuclear cells" refers to the
entire repetoir of white blood cells present in a blood sample. In a healthy
human
being, the white blood cells comprise a mixture of hematopoietic lineages
committed
and differentiated cells (typically over 99 % of the mononuclear cells are
lineages
committed cells) including, for example: Lineage committed progenitor cells
CD34+CD33+ (myeloid committed cells), CD34+CD3+ (lymphoid committed cells)
CD34+CD41+ (megakaryocytic committed cells) and differentiated cells - CD34-
CD33+ (myeloids, such as granulocytes and monocytes), CD34-CD3+, CD34-CD19+
(T and B cells, respectively), CD34-CD41+ (megakaryocytes), and hematopoietic
stem
and early progenitor cells such as CD34+Lineage negative (Liri ), CD34-Lineage
negative CD34+CD38- (typically less than 1 %).
The phrase "hematopoietic mononuclear cells which comprise a major fraction
of hematopoietic committed cells and a minor fraction of hematopoietic stem
and
progenitor cells" is used herein to describe any portion of the white blood
cells
fraction, in which the majority of the cells are hematopoietic committed
cells, while
the minority of the cells are hematopoietic stem and progenitor cells, as
these terms
are further defined hereinunder.
Hematopoietic mononuclear cells are typically obtained from a blood sample
by applying the blood sample onto a Ficoll-Hypaque layer and collecting,
following
density-cussion centrifugation, the interface layer present between the Ficoll-
Hypaque
and the blood serum, which interface layer essentially entirely consists of
the white
blood cells present in the blood sample.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
34
As used herein, the phrase "hematopoietic committed cells" refers to
differentiated hematopoietic cells that are committed to a certain
hematopoietic cell
lineage and hence can develop under physiological conditions substantially
only to
this specific hematopoietic lineage.
As used herein, the phrase "hematopoietic stem cells" refers to pluripotent
hematopoietic cells that, given the right growth conditions, may develop to
any cell
lineage present blood. This phrase, as used herein, refers both to the
earliest
renewable hematopoietic cell populations responsible for generating cell mass
in the
blood (e.g., CD34-/AC133+, CD34-/AC133-/Lineage , CD34+/AC133+ cells) and the
t 0 very early hematopoietic progenitor cells, which are somewhat more
differentiated,
yet are not committed and can readily revert to become a part of the earliest
renewable hematopoietic cell population (e.g., CD34+ cells, especially
CD34+CD38-
cells).
In normal human, most of the hematopoietic pluripotent stem cells and the
lineage committed progenitor cells are CD34+. The majority of cells are
CD34+CD38+, with a minority of cells (< 10 %) being CD34+CD38-.
The CD34+CD38- stem cells fraction identifies the most immature
hematopoietic cells, which are capable of self renewal and multilineage
differentiation. This fraction contains more long-term culture initiating
cells (LTC-
2o IC) pre-CFU and exhibits longer maintenance of the stem cells and delayed
proliferative response to cytokines as compared with the CD34+CD38+ cell
fraction.
Presently, hematopoietic stem cells are obtained by further enrichment of the
hematopoietic mononuclear cells obtained by differential density
centrifugation as
described above. This further enrichment process is typically performed by
immuno
separation such as immunomagnetic-separation or FAGS and results in a cell
fraction
that is enriched for hematopoietic stem cells.
Hence, using hematopoietic mononuclear cells as a direct source for obtaining
expanded population of hematopoietic stem cells circumvents the need for stem
cell
enrichment prior to expansion, thereby substantially simplifying the process
in terms
of both efficiency and cost.
As used herein the term "inhibiting" refers to slowing, decreasing, delaying,
preventing or abolishing.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
As used herein the term "differentiation" refers to relatively generalized or
specialized changes during development. Cell differentiation of various
lineages is a
well-documented process and requires no further description herein. As used
herein
the term differentiation is distinct from maturation which is a process,
although some
5 times associated with cell division, in which a specific cell type mature to
function
and then dies, e.g., via programmed cell death.
The phrase "cell expansion" is used herein to describe a process of cell
proliferation substantially devoid of cell differentiation. Cells that undergo
expansion
hence maintain their cell renewal properties and are oftentimes referred to
herein as
1o renewable cells, e.g., renewable stem cells.
Expansion of hematopoietic stem cells using hematopoietic mononuclear cells
as a source for the hematopoietic stem cells, as taught by the present
invention,
therefore result in converting the minor fraction (of less than 1 %) of
hematopoietic
stem and progenitor cells present in the mononuclear cells into at least the
major, if
~ 5 not the sole hematopoietic cells population post expansion, whereby in the
course of
stem cells expansion the committed cells are either substantially diluted
and/or die.
As used herein the term "ex-vivo" refers to a process in which cells are
removed from a living organism and are propagated outside the organism (e.g.,
in a
test tube). As used herein, the term "ex-vivo", however, does not refer to a
process by
20 which cells known to propagate only in-vitro, such as various cell lines
(e.g., HL-60,
MEL, HeLa, etc.) are cultured. In other words, cells expanded ex-vivo
according to
the present invention do not transform into cell lines in that they eventually
undergo
differentiation.
Providing the ex-vivo grown cells with conditions for ex-vivo cell
proliferation
25 include providing the cells with nutrients and preferably with one or more
cytokines,
as is further detailed hereinunder.
As mentioned hereinabove, concomitant with treating the hematopoietic
mononuclear cells with conditions which allow the cells to proliferate ex-
vivo, the
cells are short-term treated or long-term treated to reduce the expression
and/or
30 activity of CD38.
In one embodiment of the present invention, reducing the activity of CD38 is
effected by providing the cells with an agent that inhibits CD38 activity
(i.e., a CD38
inhibitor).



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
36
As used herein a "CD38 inhibitor" refers to an agent which is capable of
downregulating or suppressing CD38 activity in stem cells.
A CD38 inhibitor according to this aspect of the present invention can be a
"direct inhibitor" which inhibits CD38 intrinsic activity or an "indirect
inhibitor"
which inhibits the activity or expression of CD38 signaling components (e.g.,
the
cADPR and ryanodine signaling pathways) or other signaling pathways which are
effected by CD38 activity.
According to presently known embodiments of this aspect of the present
invention, nicotinamide is a possible CD38 inhibitor.
Hence, in one embodiment, the method according to this aspect of the present
invention is effected by providing the hematopoietic mononuclear cells either
with
nicotinamide itself, or with a nicotinamide analog, a nicotinamide or a
nicotinamide
analog derivative or a nicotinamide or a nicotinamide analog metabolite.
As used herein, the phrase "nicotinamide analog" refers to any molecule that
is
known to act similarly to nicotinamide. Representative examples of
nicotinamide
analogs include, without limitation, benzamide, nicotinethioamide (the thiol
analog of
nicotinamide), nicotinic acid and a-amino-3-indolepropionic acid.
The phrase "a nicotinamide or a nicotinamide analog derivative" refers to any
structural derivative of nicotinamide itself or of an analog of nicotinamide.
Examples
of such derivatives include, without limitation, substituted benzamides,
substituted
nicotinamides and nicotinethioamides and N-substituted nicotinamides and
nicotinthioamides.
The phrase "a nicotinamide or a nicotinamide analog metabolite" refers to
products that are derived from nicotinamide or from analogs thereof such as,
for
example, NAD, NADH and NADPH.
Alternatively, a CD38 inhibitor according to this aspect of the present
invention can be an activity-neutralizing antibody that binds, for example, to
the
CD38 catalytic domain, thereby inhibiting CD38 catalytic activity. It will be
appreciated, though, that since CD38 is an intracellular protein measures are
taken to
3o use inhibitors which may be delivered through the plasma membrane. In this
respect
a fragmented antibody such as a Fab fragment (described hereinunder) is
preferably
used.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
37
The term "antibody" as used in this invention includes intact molecules as
well
as functional fragments thereof, such as Fab, F(ab')2, and Fv that are capable
of
binding to macrophages. These functional antibody fragments are defined as
follows:
Fab, the fragment which contains a monovalent antigen-binding fragment of
an antibody molecule, can be produced by digestion of whole antibody with the
enzyme papain to yield an intact light chain and a portion of one heavy chain;
Fab', the fragment of an antibody molecule that can be obtained by treating
whole antibody with pepsin, followed by reduction, to yield an intact light
chain and a
portion of the heavy chain; two Fab' fragments are obtained per antibody
molecule;
(Fab')Z, the fragment of the antibody that can be obtained by treating whole
antibody with the enzyme pepsin without subsequent reduction; F(ab')Z is a
dimer of
two Fab' fragments held together by two disulfide bonds;
Fv, defined as a genetically engineered fragment containing the variable
region of the light chain and the variable region of the heavy chain expressed
as two
chains; and
Single chain antibody ("SCA"), a genetically engineered molecule containing
the variable region of the light chain and the variable region of the heavy
chain, linked
by a suitable polypeptide linker as a genetically fused single chain molecule.
Methods of making these fragments are known in the art. (See for example,
Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor
Laboratory,
New York, 1988, incorporated herein by reference).
Antibody fragments according to the present invention can be prepared by
expression in E. coli or mammalian cells (e.g. Chinese hamster ovary cell
culture or
other protein expression systems) of DNA encoding the fragment.
Antibody fragments can be obtained by pepsin or papain digestion of whole
antibodies by conventional methods. For example, antibody fragments can be
produced by enzymatic cleavage of antibodies with pepsin to provide a 5S
fragment
denoted F(ab')2. This fragment can be further cleaved using a thiol reducing
agent,
and optionally a blocking group for the sulfhydryl groups resulting from
cleavage of
disulfide linkages, to produce 3.SS Fab' monovalent fragments. Alternatively,
an
enzymatic cleavage using pepsin produces two monovalent Fab' fragments and an
Fc
fragment directly. These methods are described, for example, by Goldenberg,
U.S.
Pat. Nos. 4,036,945 and 4,331,647, and references contained therein, which
patents



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
38
are hereby incorporated by reference in their entirety. See also Porter, R.
R.,
Biochem. J., 73: 119-126, 1959. Other methods of cleaving antibodies, such as
separation of heavy chains to form monovalent light-heavy chain fragments,
further
cleavage of fragments, or other enzymatic, chemical, or genetic techniques may
also
be used, so long as the fragments bind to the antigen that is recognized by
the intact
antibody.
Fv fragments comprise an association of VE, and VL chains. This association
may be noncovalent, as described in mbar et al., Proc. Nat'1 Acad. Sci. USA
69:2659-
62, 1972. Alternatively, the variable chains can be linked by an
intermolecular
disulfide bond or cross-linked by chemicals such as glutaraldehyde.
Preferably, the
Fv fragments comprise VH and VL chains connected by a peptide linker. These
single-chain antigen binding proteins (sFv) are prepared by constructing a
structural
gene comprising DNA sequences encoding the VH and VL domains connected by an
oligonucleotide. The structural gene is inserted into an expression vector,
which is
subsequently introduced into a host cell such as E. coli. The recombinant host
cells
synthesize a single polypeptide chain with a linker peptide bridging the two V
domains. Methods for producing sFvs are described, for example, by Whitlow and
Filpula, Methods, 2: 97-105, 1991; Bird et al., Science 242:423-426, 1988;
Pack et al.,
Bio/Technology 11:1271-77, 1993; and Ladner et al., U.S. Pat. No. 4,946,778,
which
is hereby incorporated by reference in its entirety.
Another form of an antibody fragment is a peptide coding for a single
complementarity-determining region (CDR). CDR peptides ("minimal recognition
units") can be obtained by constructing genes encoding the CDR of an antibody
of
interest. Such genes are prepared, for example, by using the polymerise chain
reaction to synthesize the variable region from RNA of antibody-producing
cells.
See, for example, Larrick and Fry, Methods, 2: 106-10, 1991.
Humanized forms of non-human (e.g., murine) antibodies are chimeric
molecules of immunoglobulins, immunoglobulin chains or fragments thereof (such
as
Fv, Fab, Fab', F(ab')2 or other antigen-binding subsequences of antibodies)
which
contain minimal sequence derived from non-human immunoglobulin. Humanized
antibodies include human immunoglobulins recipient antibody in which residues
form
a complementary determining region (CDR) of the recipient are replaced by
residues
from a CDR of a non-human species (donor antibody) such as mouse, rat or
rabbit



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
39
having the desired specificity, affinity and capacity. In some instances, Fv
framework
residues of the human immunoglobulin are replaced by corresponding non-human
residues. Humanized antibodies may also comprise residues which are found
neither
in the recipient antibody nor in the imported CDR or framework sequences. In
general, the humanized antibody will comprise substantially all of at least
one, and
typically two, variable domains, in which all or substantially all of the CDR
regions
correspond to those of a non-human immunoglobulin and all or substantially all
of the
FR regions are those of a human immunoglobulin consensus sequence. The
humanized antibody optimally also will comprise at least a portion of an
immunoglobulin constant region (Fc), typically that of a human immunoglobulin
[Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature, 332:323-
329
(1988); and Presta, Curr. Op. Struct. Biol., 2:593-596 (1992)].
Methods for humanizing non-human antibodies are well known in the art.
Generally, a humanized antibody has one or more amino acid residues introduced
into
it from a source that is non-human. These non-human amino acid residues are
often
referred to as import residues, which are typically taken from an import
variable
domain. Humanization can be essentially performed following the method of
Winter
and co-workers [Jones et al., Nature, 321:522-525 (1986); Riechmann et al.,
Nature
332:323-327 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988)], by
substituting rodent CDRs or CDR sequences for the corresponding sequences of a
human antibody. Accordingly, such humanized antibodies are chimeric antibodies
(U.S. Pat. No. 4,816,567), wherein substantially less than an intact human
variable
domain has been substituted by the corresponding sequence from a non-human
species. In practice, humanized antibodies are typically human antibodies in
which
some CDR residues and possibly some FR residues are substituted by residues
from
analogous sites in rodent antibodies.
Human antibodies can also be produced using various techniques known in the
art, including phage display libraries (Hoogenboom and Winter, J. Mol. Biol.,
227:381 ( 1991 ); Marks et al., J. Mol. Biol., 222:581 ( 1991 )). The
techniques of Cole
3o et al. and Boerner et al. are also available for the preparation of human
monoclonal
antibodies (Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R.
Liss, p.
77 (1985) and Boerner et al., J. Immunol., 147(1):86-95 (1991)]. Similarly,
human
can be made by introducing of human immunoglobulin loci into transgenic
animals,



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
e.g., mice in which the endogenous immunoglobulin genes have been partially or
completely inactivated. Upon challenge, human antibody production is observed,
which closely resembles that seen in humans in all respects, including gene
rearrangement, assembly, and antibody repertoire. This approach is described,
for
5 example, in U.S. Pat. Nos. 5,545,807; 5,545,806; 5,569,825; 5,625,126;
5,633,425;
5,661,016, and in the following scientific publications: Marks et al.,
Bio/Technology
10, 779-783 (1992); Lonberg et al., Nature 368 856-859 (1994); Morrison,
Nature 368
812-13 (1994); Fishwild et al., Nature Biotechnology 14, 845-S1 (1996);
Neuberger,
Nature Biotechnology 14, 826 (1996); Lonberg and Huszar, Intern. Rev. Immunol.
13
10 65-93 ( 1995).
Alternatively, the method according to this aspect of the present invention
can
be effected by providing the ex-vivo cultured hematopoietic mononuclear cells
with
an agent that downregulates CD38 expression.
An agent that downregulates CD38 expression refers to any agent which
15 affects CD38 synthesis (decelerates) or degradation (accelerates) either at
the level of
the mRNA or at the level of the protein. For example, a small interfering
polynucleotide molecule which is designed to down regulate the expression of
CD38
can be used according to this aspect of the present invention.
An example for a small interfering polynucleotide molecule which can down-
20 regulate the expression of CD38 is a small interfering RNA or siRNA, such
as, for
example, the morpholino antisense oligonucleotides described by in Munshi et
al.
(Munshi CB, Graeff R, Lee HC, JBiol Chem 2002 Dec 20;277(51):49453-8), which
includes duplex oligonucleotides which direct sequence specific degradation of
mRNA through the previously described mechanism of RNA interference (RNAi)
25 (Hutvagner and Zamore (2002) Curr. Opin. Genetics and Development 12:225-
232).
As used herein, the phrase "duplex oligonucleotide" refers to an
oligonucleotide structure or mimetics thereof, which is formed by either a
single self
complementary nucleic acid strand or by at least two complementary nucleic
acid
strands. The "duplex oligonucleotide" of the present invention can be composed
of
3o double-stranded RNA (dsRNA), a DNA-RNA hybrid, single-stranded RNA (ssRNA),
isolated RNA (i.e., partially purified RNA, essentially pure RNA), synthetic
RNA and
recombinantly produced RNA.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
41
Preferably, the specific small interfering duplex oligonucleotide of the
present
invention is an oligoribonucleotide composed mainly of ribonucleic acids.
Instructions for generation of duplex oligonucleotides capable of mediating
RNA interference are provided in www.ambion.com.
s Hence, the small interfering polynucleotide molecule according to the
present
invention can be an RNAi molecule (RNA interference molecule).
Alternatively, a small interfering polynucleotide molecule can be an
oligonucleotide such as a CD38-specific antisense molecule or a rybozyme
molecule,
further described hereinunder.
Oligonucleotides designed according to the teachings of the present invention
can be generated according to any oligonucleotide synthesis method known in
the art
such as enzymatic synthesis or solid phase synthesis. Equipment and reagents
for
executing solid-phase synthesis are commercially available from, for example,
Applied Biosystems. Any other means for such synthesis may also be employed;
the
actual synthesis of the oligonucleotides is well within the capabilities of
one skilled in
the art.
Oligonucleotides used according to this embodiment of the present invention
are those having a length selected from a range of 10 to about 200 bases
preferably
15-150 bases, more preferably 20-100 bases, most preferably 20-50 bases.
The oligonucleotides of the present invention may comprise heterocyclic
nucleosides consisting of purines and the pyrimidines bases, bonded in a 3' to
5'
phosphodiester linkage.
Preferably used oligonucleotides are those modified in either backbone,
internucleoside linkages or bases, as is broadly described hereinunder. Such
modifications can oftentimes facilitate oligonucleotide uptake and resistivity
to
intracellular conditions.
Specific examples of preferred oligonucleotides useful according to this
aspect
of the present invention include oligonucleotides containing modified
backbones or
non-natural internucleoside linkages. Oligonucleotides having modified
backbones
include those that retain a phosphorus atom in the backbone, as disclosed in
U.S.
Patents Nos.: 4,687,808; 4,469,863; 4,476,301; 5,023,243; 5,177,196;
5,188,897;
5,264,423; 5,276,019; 5,278,302; 5,286,717; 5,321,131; 5,399,676; 5,405,939;



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
42
5,453,496; 5,455,233; 5,466, 677; 5,476,925; 5,519,126; 5,536,821; 5,541,306;
5,550,111; 5,563,253; 5,571,799; 5,587,361; and 5,625,050.
Preferred modified oligonucleotide backbones include, for example,
phosphorothioates, chiral phosphorothioates, phosphorodithioates,
phosphotriesters,
aminoalkyl phosphotriesters, methyl and other alkyl phosphonates including 3'
alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates
including 3'-amino phosphoramidate and aminoalkylphosphoramidates,
thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters,
and
boranophosphates having normal 3'-5' linkages, 2'-5' linked analogs of these,
and
those having inverted polarity wherein the adjacent pairs of nucleoside units
are
linked 3'-5' to 5'-3' or 2'-5' to 5'-2'. Various salts, mixed salts and free
acid forms can
also be used.
Alternatively, modified oligonucleotide backbones that do not include a
phosphorus atom therein have backbones that are formed by short chain alkyl or
cycloalkyl internucleoside linkages, mixed heteroatom and alkyl or cycloalkyl
internucleoside linkages, or one or more short chain heteroatomic or
heterocyclic
internucleoside linkages. These include those having morpholino linkages
(formed in
part from the sugar portion of a nucleoside); siloxane backbones; sulfide,
sulfoxide
and sulfone backbones; formacetyl and thioformacetyl backbones; methylene
formacetyl and thioformacetyl backbones; alkene containing backbones;
sulfamate
backbones; methyleneimino and methylenehydrazino backbones; sulfonate and
sulfonamide backbones; amide backbones; and others having mixed N, O, S and
CHZ
component parts, as disclosed in U.S. Pat. Nos. 5,034,506; 5,166,315;
5,185,444;
5,214,134; 5,216,141; 5,235,033; 5,264,562; 5,264,564; 5,405,938; 5,434,257;
5,466,677; 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240;
5,610,289; 5,602,240; 5,608,046; 5,610,289; 5,618,704; 5,623, 070; 5,663,312;
5,633,360; 5,677,437; and 5,677,439.
Other oligonucleotides which can be used according to the present invention,
are those modified in both sugar and the internucleoside linkage, i.e., the
backbone, of
the nucleotide units are replaced with novel groups. The base units are
maintained for
complementation with the appropriate polynucleotide target. An example for
such an
oligonucleotide mimetic, includes peptide nucleic acid (PNA). A PNA
oligonucleotide refers to an oligonucleotide where the sugar-backbone is
replaced



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
43
with an amide containing backbone, in particular an aminoethylglycine
backbone. The
bases are retained and are bound directly or indirectly to aza nitrogen atoms
of the
amide portion of the backbone. United States patents that teach the
preparation of
PNA compounds include, but are not limited to, U.S. Pat. Nos. 5,539,082;
5,714,331;
and 5,719,262, each of which is herein incorporated by reference. Other
backbone
modifications, which can be used in the present invention are disclosed in
U.S. Pat.
No: 6,303,374.
Oligonucleotides of the present invention may also include base modifications
or substitutions. As used herein, "unmodified" or "natural" bases include the
purine
bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T),
cytosine
(C) and uracil (U). Modified bases include but are not limited to other
synthetic and
natural bases such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine,
xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives
of
adenine and guanine, 2-propyl and other alkyl derivatives of adenine and
guanine, 2-
thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-
propynyl
uracil and cytosine, 6-azo uracil, cytosine and thymine, S-uracil
(pseudouracil), 4-
thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-
substituted
adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and
other 5-
substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 8-
azaguanine and 8-azaadenine, 7-deazaguanine and 7-deazaadenine and 3-
deazaguanine and 3-deazaadenine. Further bases include those disclosed in U.S.
Pat.
No: 3,687,808, those disclosed in The Concise Encyclopedia Of Polymer Science
And
Engineering, pages 858-859, Kroschwitz, J. L, ed. John Wiley & Sons, 1990,
those
disclosed by Englisch et al., Angewandte Chemie, International Edition, 1991,
30,
613, and those disclosed by Sanghvi, Y. S., Chapter 15, Antisense Research and
Applications, pages 289-302, Crooke, S. T. and Lebleu, B., ed., CRC Press,
1993.
Such bases are particularly useful for increasing the binding affinity of the
oligomeric
compounds of the invention. These include 5-substituted pyrimidines, 6-
azapyrimidines and N-2, N-6 and O-6 substituted purines, including 2-
aminopropyladenine, 5-propynyluracil and 5-propynylcytosine. 5-methylcytosine
substitutions have been shown to increase nucleic acid duplex stability by 0.6-
1.2 °C.
[Sanghvi YS et al. (1993) Antisense Research and Applications, CRC Press, Boca



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
44
Raton 276-278] and are presently preferred base substitutions, even more
particularly
when combined with 2'-O-methoxyethyl sugar modifications.
Another modification of the oligonucleotides of the invention involves
chemically linking to the oligonucleotide one or more moieties or conjugates,
which
enhance the activity, cellular distribution or cellular uptake of the
oligonucleotide.
Such moieties include but are not limited to lipid moieties such as a
cholesterol
moiety, cholic acid, a thioether, e.g., hexyl-S-tritylthiol, a
thiocholesterol, an aliphatic
chain, e.g., dodecandiol or undecyl residues, a phospholipid, e.g., di-
hexadecyl-rac-
glycerol or triethylammonium 1,2-di-O-hexadecyl-rac-glycero-3-H-phosphonate, a
l0 polyamine or a polyethylene glycol chain, or adamantine acetic acid, a
palmityl
moiety, or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety, as
disclosed in U.S. Pat. No: 6,303,374.
It is not necessary for all positions in a given oligonucleotide molecule to
be
uniformly modified, and in fact more than one of the aforementioned
modifications
may be incorporated in a single compound or even at a single nucleoside within
an
oligonucleotide.
As described hereinabove, the oligonucleotides of the present invention are
preferably antisense molecules, which are chimeric molecules. "Chimeric
antisense
molecules" are oligonucleotides, which contain two or more chemically distinct
regions, each made up of at least one nucleotide. These oligonucleotides
typically
contain at least one region wherein the oligonucleotide is modified so as to
confer
upon the oligonucleotide increased resistance to nuclease degradation,
increased
cellular uptake, and/or increased binding affinity for the target
polynucleotide. An
additional region of the oligonucleotide may serve as a substrate for enzymes
capable
of cleaving RNA:DNA or RNA:RNA hybrids. An example for such includes RNase
H, which is a cellular endonuclease which cleaves the RNA strand of an RNA:DNA
duplex. Activation of RNase H, therefore, results in cleavage of the RNA
target,
thereby greatly enhancing the efficiency of oligonucleotide inhibition of gene
expression. Consequently, comparable results can often be obtained with
shorter
oligonucleotides when chimeric oligonucleotides are used, compared to
phosphorothioate deoxyoligonucleotides hybridizing to the same target region.
Cleavage of the RNA target can be routinely detected by gel electrophoresis
and, if
necessary, associated nucleic acid hybridization techniques known in the art.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
Chimeric antisense molecules of the present invention may be formed as
composite structures of two or more oligonucleotides, modified
oligonucleotides, as
described above. Representative U.S. patents that teach the preparation of
such
hybrid structures include, but are not limited to, U.S. Pat. Nos. 5,013,830;
5,149,797;
5 5,220,007; 5,256,775; 5,366,878; 5,403,711; 5,491,133; 5,565,350; 5,623,065;
5,652,355; 5,652,356; and 5,700,922, each of which is herein fully
incorporated by
reference.
The oligonucleotides of the present invention can further comprise a ribozyme
sequence. Ribozymes are being increasingly used for the sequence-specific
inhibition
10 of gene expression by the cleavage of mRNAs. Several rybozyme sequences can
be
fused to the oligonucleotides of the present invention. These sequences
include but
are not limited ANGIOZYME specifically inhibiting formation of the VEGF-R
(Vascular Endothelial Growth Factor receptor), a key component in the
angiogenesis
pathway, and HEPTAZYME, a rybozyme designed to selectively destroy Hepatitis C
15 Virus (HCV) RNA, (Rybozyme Pharmaceuticals, Incorporated - WEB home page).
Further alternatively, a small interfering polynucleotide molecule, according
to
the present invention can be a DNAzyme.
DNAzymes are single-stranded catalytic nucleic acid molecules. A general
model (the "10-23" model) for the DNAzyme has been proposed. "10-23"
20 DNAzymes have a catalytic domain of 15 deoxyribonucleotides, flanked by two
substrate-recognition domains of seven to nine deoxyribonucleotides each. This
type
of DNAzyme can effectively cleave its substrate RNA at purine:pyrimidine
junctions
(Santoro, S.W. & Joyce, G.F. Proc. Natl, Acad. Sci. USA 199; for rev of
DNAzymes
see Khachigian, LM Curr Opin Mol Ther 2002;4:119-21 ).
25 Examples of construction and amplification of synthetic, engineered
DNAzymes recognizing single and double-stranded target cleavage sites have
been
disclosed in U.S. Pat. No. 6,326,174 to Joyce et al. DNAzymes of similar
design
directed against the human Urokinase receptor were recently observed to
inhibit
Urokinase receptor expression, and successfully inhibit colon cancer cell
metastasis in
30 vivo (Itoh et al., 20002, Abstract 409, Ann Meeting Am Soc Gen Ther
www.as~t.or~). In another application, DNAzymes complementary to bcr-abl
oncogenes were successful in inhibiting the oncogenes expression in leukemia
cells,



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
46
and lessening relapse rates in autologous bone marrow transplant in cases of
CML
and ALL.
Alternatively, as mentioned hereinabove and is further detailed in
PCT/IL03/00064 and U.S. Provisional Patent Application No. 60/452,545,
retinoid
receptor superfamily inhibitors (e.g., antagonists, siRNA molecules, antisense
molecules, antibodies, etc.) which downregulate or suppress retinoid receptor
activity
and/or expression can be used to downregulate CD38 expression.
Briefly, retinoid receptors such as retinoic acid receptor (RAR), retinoid X
receptor (RXR) and vitamin D receptor (VDR) have been reported to be involved
in
l0 the regulation of gene expression pathways associated with cell
proliferation and
differentiation and in particular in the regulation of CD38 expression [Kapil
M.,
Teresa M., Taghi M., Michael A., Steven C., Maher A.. Involvement of retinoic
acid
receptor mediated signaling pathway in induction of CD38 cell surface antigen,
Blood.
1997; 89:3607-3614; Ueno H, Kizaki M, Matsushita H, Muto A, Yamato K,
Nishihara
T, Hida T, Yoshimura H, Koeffler HP, Ikeda Y. A novel retinoic acid receptor
(RAR)-
selective antagonist inhibits differentiation and apoptosis of HL-60 cells:
implications
of RAR alpha-mediated signals in myeloid leukemic cells. Leuk Res. 1998;
22:517-
25]. Hence, preferred agents that downregulate CD38 expression according to
the
present invention include RAR antagonists, RXR antagonists and VDR antagonists
or,
alternatively, antagonists for reducing the capacity of the hematopoietic
mononuclear
cells in responding to retinoic acid, retinoid and/or Vitamin D.
As used herein the term "antagonist" refers to an agent that counteracts or
abrogates the effects of an agonist or a natural ligand of a receptor. Further
features
relating to such antagonists are detailed hereinunder.
Further alternatively, as is described in detail in U.S. Provisional Patent
Application No. 60/452,545, down regulation of CD38 expression can be obtained
by
downregulating the expression and/or activity of phosphatidyl inositol 3-
kinase,
which is also referred to herein throughout as PI 3-kinase. Briefly, it has
been
reported that PI 3-kinase plays a critical function in the activation of
nuclear receptors
such as the retinoid receptor superfamily and the vitamin D receptor, as an
obligatory
factor for proper receptor signaling pathways and is hence involved in cell
differentiation.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
47
Hence, agents that interfere with PI 3-kinase expression and/or activity are
also preferred agents for downregulating CD38, according to the present
invention.
Representative examples of agents that inhibit PI 3-kinase activity include,
but are not
limited to, the known PI 3-kinase inhibitors wortmannin and LY294002, and
analogs,
derivatives, and metabolites thereof. Additional examples of PI 3-kinase
inhibitors
are described in U.S. Patent No. 5,378,725, which is incorporated by reference
as if
fully set forth herein. Representative examples of agents that downregulate PI
3-
kinase expression according to the present invention include, but are not
limited to,
polynucleotides, such as small interfering RNA molecules, antisense ribozymes
and
DNAzymes, as well as intracellular antibodies, using the methodologies
described
hereinabove with respect to downregulating CD38 expression.
Each of the agents described hereinabove may reduce the expression or
activity of CD38 individually. However, the present invention aims to also
encompass the use of any subcombination of these agents.
It will be appreciated that protein agents (e.g., antibodies) of the present
invention can be expressed from a polynucleotide encoding same and provided to
ex-
vivo cultured hematopoietic mononuclear cells employing an appropriate gene
delivery vehicle/method and a nucleic acid construct as is further described
hereinunder.
2o Examples of suitable constructs include, but are not limited to pcDNA3,
pcDNA3.l (+/-), pGL3, PzeoSV2 (+/-), pDisplay, pEF/myc/cyto, pCMV/myc/cyto
each of which is commercially available from Invitrogen Co.
(www.invitrogen.com).
Examples of retroviral vector and packaging systems are those sold by
Clontech, San
Diego, Cali~, including Retro-X vectors pLNCX and pLXSN, which permit cloning
into multiple cloning sites and the transgene is transcribed from CMV
promoter.
Vectors derived from Mo-MuLV are also included such as pBabe, where the
transgene will be transcribed from the 5'LTR promoter.
As the method of ex-vivo expanding a population of hematopoietic stem cells,
according to this aspect of the present invention, is effected by modulating
CD38
expression and/or activity, either at the protein level, using RAR, RXR or VDR
antagonists, a PI-3 kinase inhibitor or a CD38 inhibitor such as nicotinamide
and
analogs thereof, or at the at the expression level via genetic engineering
techniques, as
is detailed hereinabove, there are further provided, according to the present
invention,



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
48
several preferred methods of ex-vivo expanding a population of hematopoietic
stem
cells of hematopoietic mononuclear cells.
In one particular, a method of ex-vivo expanding a population of
hematopoietic stem cells, while at the same time, substantially inhibiting
differentiation of the hematopoietic stem cells ex-vivo is effected by
providing
hematopoietic mononuclear cells which comprise a major fraction of
hematopoietic
committed cells and a minor fraction of hematopoietic stem and progenitor
cells, with
ex-vivo culture conditions for ex-vivo cell proliferation and, at the same
time, for
reducing a capacity of the hematopoietic mononuclear cells in responding to
retinoic
acid, retinoids and/or Vitamin D, so as to expand a population of
hematopoietic stem
cells, while at the same time, substantially inhibiting differentiation of the
hematopoietic stem cells ex-vivo.
Reducing the capacity of the cells in responding to retinoic acid, retinoids
and/or Vitamin D, or to retinoic acid, retinoid X and/or Vitamin D receptor
signaling
~ 5 may be effected, for example, by the administration of chemical
inhibitors, including
receptor antagonists.
In another particular, the method of ex-vivo expanding a population of stem
cells, while at the same time, substantially inhibiting differentiation of the
stem cells
ex-vivo is effected by providing hematopoietic mononuclear cells which
comprise a
major fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem and progenitor cells, with ex-vivo culture conditions for
ex-vivo
cell proliferation and, at the same time, for reducing a capacity of the
hematopoietic
mononuclear cells in responding to signaling pathways involving the retinoic
acid
receptor, retinoid-X receptor and/or Vitamin D receptor, to thereby expand a
population of hematopoietic stem cells, while at the same time, substantially
inhibiting differentiation of the hematopoietic stem cells ex-vivo.
Reducing the capacity of the cells to respond to retinoic acid, retinoid X
and/or
Vitamin D receptor signaling events, includes treating the cells with
antagonists
supplied continuously or for a short-pulse period, and is effected by a
diminution or
abrogation of cellular signaling pathways through their respective, cognate
receptors.
As is described and exemplified in PCT/ILO1/00064, reducing the capacity of
hematopoietic cells in responding to the disclosed signaling pathways is
reversible,
e.g., inherently reversible. In other words, cells expanded using the
protocols of the



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
49
present invention do not transform into cell lines. Hence, by exposing the
cells
following sufficient expansion to growth conditions by which differentiation
is
induced, one would be able to direct the ex-vivo differentiation of the cells
to desired
direction, including ex-vivo and in vivo cis- and trans-differentiation.
As used herein "cis-differentiation" refers to differentiation of adult stem
cells
into a tissue from which they were derived. For example, the differentiation
of
CD34+ hematopoietic cells to different committed/mature blood cells
constitutes cis-
differentiation.
As used herein "trans-differentiation" refers to differentiation of adult stem
cells into a tissue from which they were not derived. For example, the
differentiation
of CD34+ hematopoietic cells to cells of different tissue origin, e.g.,
myocites
constitutes trans-differentiation.
Reducing the capacity of the hematopoietic mononuclear cells in responding to
the above antagonists and/or signaling pathways of the above receptors and
kinase is
IS effected by ex-vivo culturing hematopoietic mononuclear cells in a presence
of an
effective amount of at least one retinoic acid receptor antagonist, at least
one retinoid
X receptor antagonist and/or at least one Vitamin D receptor antagonist,
preferably,
for a time period of 0.1-50 %, preferably, 0.1-25 %, more preferably, 0.1-15
%, of an
entire ex-vivo culturing period of the hematopoietic mononuclear cells or for
the entire
period. In this respect it was surprisingly uncovered that an initial pulse
exposure to
an antagonist is sufficient to exert cell expansion long after the antagonist
was
removed from the culturing set up.
Final concentrations of the antagonists may be, depending on the specific
application, in the micromolar or millimolar ranges. For example, within about
0.1 p
M to about 100 mM, preferably within about 4 ~M to about 50 mM, more
preferably
within about S yM to about 40 mM.
Many antagonists to RAR, RXR and VDR, which are usable in this and other
aspects and embodiments of the present invention, are presently known.
Representative examples of such retinoic acid receptor antagonist include,
3o without limitation, AGN 194310; AGN 109; 3-(4-Methoxy-phenylsulfanyl)-3-
methyl-butyric acid; 6-Methoxy-2,2-dimethvl-thiochroman-4-one,2,2-Dimethyl-4-
oxo-thiochroman-6-yltrifluoromethane-sulfonate; Ethyl 4-((2,2 dimethyl-4-oxo-
thiochroman-6-yl)ethynyl)-benzoate; Ethyl 4-((2,2-dimethy 1-4-



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
triflouromethanensulfonyloxy -(2H)- thiochromen-6-yl)ethynyl)-benzoate(41);
Thiochromen-6-yl]-ethynyl]-benzoate(yl); (p-[(E)-2-[3'4'-Dihydro-4,4'-dimethyl-
7'-
(heptyloxy)-2'H-1-benzothiopyran-6'yl] propenyl] benzoic acid 1'1'-dioxide;
2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-butoxyphenyl)-3-methyl]-octa-2,4,6-trienoic
acid;
5 2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-propoxyphenyl)-3-methyl]-octa-2,4,6-trienoic
acid;
2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-pentoxyphenyl)-3-methyl]-octa-2,4,6-trienoic
acid;
2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-hexoxyphenyl)-3-methyl]-octa-2,4,6-trienoic
acid;
2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-heptoxyphenyl)-3-methyl]-octa-2,4,6-trienoic
acid;
2E,4E,6E-[7-(3,5-Di-t-butyl-4-n-octoxyphenyl)-3-methyl]-octa-2,4,6-trienoic
acid;
l0 (2E,4E,6E)-7-[3-t-butyl-5-(1-phenyl-vinyl)-phenyl]-3-methyl-octa-2,4,6-
trienoic acid;
2E,4E,6E-[7-(3,5-Di-t-butyl-4-{[4,5-3 H2]-n-pentoxy}phenyl)-3-
methyl]-
octa-2,4,6-trienoic acid; (2E,4E)-(1RS,2RS)-5-[2-(3,5-di-tert.butyl-2-ethoxy-
phenyl)-
cyclopropyl]-3-methyl-penta-2,4-dienoic acid ethyl ester; (2E,4E)-(1RS,2RS)-5-
[2-
(3,5-di-tert.butyl-2-ethoxy-phenyl)-cyclopropyl]-3-methyl-penta-2,4-dienoic
acid;
15 (2E,4E)-(1RS,2RS)-5-[2-(3,5-di-tert.butyl-2-butoxy-phenyl)-cyclopropyl]-3-
methyl-
penta-2,4-dienoic acid; (2E,4E,6Z)-7-[3,5-di-tert.butyl-2-ethoxyphenyl]3-
methyl-
2,4,6-octatrienoic acid; (2E,4E,6Z)-7-[3,5-di-tert.butyl-2-butyloxyphenyl]-3-
methyl-
2,4,6-octatrienoic acid; 4-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-
naphthalene-
carboxamido) benzoic acid; (2E,4E)-3-methyl-5-[(1S,2S)-2-(5,5,8,8-tetramethyl-
20 5,6,7,8-tetrahydro-naphthalen-2-yl)-cyclopropyl]-penta-2,4-dienoic acid; p-
[(E)-2-
[3',4'-Dihydro-4',4'-dimethyl-7'-(heptyloxy)-2'H-1-benzothiopyran-6'-
yl]propenyl]benzoic acid; 1',1'-dioxide, 4-(7,7,10,10-Tetramethyl-1-pyridin-3-
ylmethyl-4,5,7,8,9,10-hexahydro-1H-naphto[2,3-g]indol-3-yl)-benzoic acid;
(2E,4E,6Z)-7-[3,5-di-tert.butyl-2-methoxyphenyl]-3-methyl-2,4,6-octatrienoic
acid;
25 (2E,4E,6Z)-7-[3,5-di-tert.butyl-2-ethoxyphenyl]-3-methyl-2,4,6-octatrienoic
acid;
(2E,4E,6Z)-7-[3,5-di-tert.butyl-2-hexyloxyphenyl]-3-methyl-2,4,6-octatrienoic
acid;
(2E,4E,6Z)-7-[3,5-di-tert.butyl-2-octyloxyphenyl]-3-methyl-2,4,6-octatrienoic
acid;
and (2E,4E)-(1RS,2RS)-5-[2-(3,5-di-tert-butyl-2-butoxy-phenyl)-cyclopropyl]-3-
methyl-penta-2,4-dienoic acid (2E,4E,6Z)-7-(3-n-propoxy-5,6,7,8-tetrahydro-
5,5,8,8-
30 tetramethylnaphthalene-2-yl)-3-methylocta-2,4,6-trienoic acid, and 4-(5H-
2,3(2,5
dimethyl-2,5-hexano)-5-n-propyldibenzo[b,e][1,4]diazepin-11-yl)benzoic acid,
and 4-
(SH-2,3-(2,5-dimethyl-2,5-hexano)-5methyl-8-nitrodibenzo [b,e] [ 1,4] diazepin-
11-
yl)benzoic acid, and 4-{[4-(4-Ethylphenyl)2,2-dimethyl-(2H)-thiochromen-6-



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
51
yl]ethynyl}benzoic acid, and 4-[4-2methyl-1,2-dicarba-closo-dodecaboran-1-yl-
phenylcarbamoyl]benzoic acid, and 4-[4,5,7,8,9,10-hexahydro-7,7,10,10-
tetramethyl-
1-(3-pyridylmethyl)-anthra[1,2-b]pyrrol-3-yl]benzoic acid, and (3-
pyridylmethyl)-]5-
thiaanthra[2,1-b]pyrrol-3-yl)benzoic acid, and (3-pyridylmethyl)-anthra[2m1-
d]pyrazol-3-yl]benzoic acid.
Representative examples of such retinoid X receptor antagonist include,
without limitation, LGN100572, 1-(3-hydroxy-5,6,7,8-tetrahydro-5,5,8,8-
tetramethylnaphthalene-2-yl)ethanone, 1-(3-propoxy-5,6,7,8-tetrahydro-5,5,8,8-
tetramethylnaphthalene-2-yl)ethanone, 3-(3-propoxy-5,6,7,8-tetrahydro-5,5,8,8-
tetramethylnaphthalene-2-yl)but-2-enenitrile, 3-(3-propoxy-5,6,7,8-tetrahydro-
5,5,8,8-tetramethylnaphthalene-2-yl)but-2-enal, (2E,4E,6E)-7-3[-propoxy-
5,6,7,8-
tetrahydro 5,5,8,8-tetramethyl-2-naphthalene-2-yl]-3-methylocta-2,4,6-trienoic
acid,
4-[3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)carbonyl] benzoic acid,
4-[1-
(3,5, 5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)ethenyl] benzoic acid, 4-

[1(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)cyclopropyl] benzoic
acid, 4-
[1-(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)ethenyl] benzenete
trazole,
2-[1-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydro-2-naphthyl) ethenyl]pyridine-5-
carboxylic acid, 2-[1-(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-
naphthyl)ethyl]pyridine-5-carboxylic acid, ethyl-2-[1-(3,5,5,8, 8-pentamethyl-
5,6,7,8-
2o tetrahydro-2-naphthyl)ethenyl]pyridine-5-carboxylate, 5-[1-3,5,5,8,8-
pentamethyl-
5,6,7,8-tetrahydro-2-naphthyl)ethenyl]pyridine-2-carboxylic acid, 2-[1-
(3,5,5,8,8-
pentamethyl-5,6,7,8-tetrahydro-2-naphthyl) cyclopropyl]pyridine-5-carboxylic
acid,
methyl 2-[ 1-(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-
naphthyl)cyclopropyl]pyridine-5-carboxylate, 4-[1-(3,5, 5,8,8-pentamethyl-
5,6,7,8-
tetrahydro-2-naphthyl)ethenyl]-N-(4-hydroxyphenyl) benzamide, 2-[1-(3,5,5,8,8-
Pentamethyl-5,6,7,8-tetrahydro-2-naphthyl) ethenyl] pyridine-5-carboxylic
acid, 2-[1-
(3,5,5,8,8-Pentamethyl-5, 6,7,8-tetrahydro-2-naphthyl)cyclopropyl]pyridine-5-
carboxylic acid, 4-[(3,5, 5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-
naphthyl)carbonyl]benzoic acid butyloxime, 4-[(3,5,5,8,8-pentamethyl-5,6,7,8-
tetrahydro-2-naphthyl) carbonyl]benzoic acid propyloxime, 4-[(3,5,5,8,8-
pentamethyl-5,6,7,8-terrahydro-2-naphthyl)carbonyl]benzoic acid cyanoimine, 4-
[(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)carbonyl]benzoic acid
allyloxime, 4-[(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-
naphthyl)carbonyl]benzoic



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
52
acid 4-(3-methylbut-2-enoic acid)oxime, and 4-[(3,5,5,8,8-pentamethyl-5,6,7,8-
tetrahydro-2-naphthyl)carbonyl]benzoic acid 1-aminoethyloxime (2E,4E,6Z)-7-(3-
n-
propoxy-5,6,7,8-tetrahydro-5,5,8,8-tetramethylnaphthalene-2-yl)-3-methylocta-
2,4,6-
trienoic acid, and 4-(5H-2,3(2,5 dimethyl-2,5-hexano)-5-n-
propyldibenzo[b,e][1,4]diazepin-11-yl)benzoic acid, and 4-(5H-2,3-(2,5-
dimethyl-
2,5-hexano)-5m.
Representative examples of such Vitamin D receptor antagonist include,
without limitation: 1 alpha, 25-(OH)-D3-26,23 lactone; 1 alpha, 25-
dihydroxyvitamin
D (3); the 25-carboxylic ester ZK159222; (23S)- 25-dehydro-1 alpha-OH-D (3);
(23R)-25-dehydro-1 alpha-OH-D (3); 1 beta, 25 (OH)z D3; 1 beta, 25(OH)2-3-epi-
D3;
(23S) 25-dehydro-1 alpha(OH) D3-26,23-lactone; (23R) 25-dehydro-1 alpha(OH)D3-
26,23-lactone and Butyl-(5Z,7E,22E-(1S,7E,22E-(1S,3R,24R)-1,3,24-trihydroxy-
26,27-cyclo-9,10-secocholesta-5,7,10( 19),22-tetraene-25-carboxylate).
The above listed antagonists are known for their high affinity towards their
respective cognate receptors. However, it may be possible for these molecules
to be
active towards other receptors.
Hence, in another particular, the method of ex-vivo expanding a population of
hematopoietic stem cells, while at the same time, substantially inhibiting
differentiation of the hematopoietic stem cells ex-vivo is effected by
providing
hematopoietic mononuclear cells which comprise a major fraction of
hematopoietic
committed cells and a minor fraction of hematopoietic stem and progenitor
cells, with
ex-vivo culture conditions for ex-vivo cell proliferation and, at the same
time, for
reducing a capacity of the hematopoietic mononuclear cells in responding to
signaling
pathways involving PI 3-kinase, to thereby expand a population of
hematopoietic
stem cells, while at the same time, substantially inhibiting differentiation
of the
hematopoietic stem cells ex-vivo.
In another particular, the method of ex-vivo expanding a population of
hematopoietic stem cells, while at the same time, substantially inhibiting
differentiation of the hematopoietic stem cells ex-vivo is effected by
providing
3o hematopoietic mononuclear cells which comprise a major fraction of
hematopoietic
committed cells and a minor fraction of hematopoietic stem and progenitor
cells, with
ex-vivo culture conditions for ex-vivo cell proliferation and with
nicotinamide, a
nicotinamide analog, a nicotinamide or a nicotinamide analog derivative or a



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
53
nicotinamide or a nicotinamide analog metabolite, to thereby expand a
population of
hematopoietic stem cells, while at the same time, substantially inhibiting
differentiation of the hematopoietic stem cells ex-vivo.
In yet another particular, the method of ex-vivo expanding a population of
hematopoietic stem cells, while at the same time, substantially inhibiting
differentiation of the hematopoietic stem cells ex-vivo is effected by
providing
hematopoietic mononuclear cells which comprise a major fraction of
hematopoietic
committed cells and a minor fraction of hematopoietic stem and progenitor
cells, with
ex-vivo culture conditions for ex-vivo cell proliferation and with a PI 3-
kinase
inhibitor, to thereby expand a population of hematopoietic stem cells, while
at the
same time, substantially inhibiting differentiation of the hematopoietic stem
cells ex-
vivo.
Final concentrations of the nicotinamide or the analogs, derivatives or
metabolites thereof and of the PI 3-kinase inhibitor are preferably, depending
on the
specific application, in the millimolar ranges. For example, within about 0.1
mM to
about 20 mM, preferably within about 1 mM to about 10 mM, more preferably
within
about 5 mM to about 10 mM.
As is described hereinabove and is further exemplified in the Examples
section that follows, expansion of the hematopoietic stem cells population
present in
2o hematopoietic mononuclear cells can also be effected in the presence of
copper
chelators or chalets. As is discussed in detail in WO 00/18885 and in
PCT/IL03/00062, addition of copper chelators or copper chelates to cells
culturing
media affects the cellular copper concentration, which in turn, affects
signaling
pathways associated with cells differentiation. According to the teachings of
WO
00/18885 and PCT/IL03/00062, addition of a copper chelate to the cells
culturing
media maintains the free copper concentration of the cells substantially
unchanged
during cell expansion, while addition of a copper chelator to the cells
culturing media
reduces the capacity of the cells in utilizing copper.
As used herein, the phrase "copper chelator" refers to a ligand that has at
least
two atoms capable of coordinating with copper or a copper ion, so as to form a
ring.
A copper chelator is free of, i.e., not complexed with, the copper ion.
Additional
features relating to chelating effects are described, for example, in
PCT/IL03/00062.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
54
As used herein throughout, the phrase "copper chelate" refers to a copper
chelator, as is defined hereinabove, which is complexed with a copper ion.
Hence, according to the present invention there is provided another method of
ex-vivo expanding a population of hematopoietic stem cells, while at the same
time,
substantially inhibiting differentiation of the hematopoietic stem cells ex-
vivo. The
method, according to this aspect of the present invention is effected by
providing
hematopoietic mononuclear cells which comprise a major fraction of
hematopoietic
committed cells and a minor fraction of hematopoietic stem and progenitor
cells, with
ex-vivo culture conditions for ex-vivo cell proliferation and with one or more
copper
1o chelator(s) or copper chelate(s), to thereby expand a population of
hematopoietic stem
cells, while at the same time, substantially inhibiting differentiation of the
hematopoietic stem cells ex-vivo.
The copper chelate or chelators of the present invention is oftentimes capable
of forming an organometallic complex with a transition metal other than
copper. As
metals other than copper are typically present in the cells (e.g., zinc) or
can be
administered to cells during therapy (e.g., platinum), it was found that
copper chelates
or chelators that can also interact with other metals are highly effective.
Representative examples of such transition metals include, without limitation,
zinc,
cobalt, nickel, iron, palladium, platinum, rhodium and ruthenium.
The copper chelates of the present invention comprise a copper ion (e.g.,
Cu+',
Cu+Z) and one or more copper chelator(s). Preferred copper chelators according
to the
present invention include polyamine molecules, which can form a cyclic complex
with the copper ion via two or more amine groups present in the polyamine.
Hence, the copper chelate or chelator used in the context of the different
aspects and embodiments of the present invention preferably includes a
polyamine
chelator, namely a polymeric chain that is substituted and/or interrupted with
1-10
amine moieties, preferably 2-8 amine moieties, more preferably 4-6 amine
moieties
and most preferably 4 amine moieties.
The phrases "amine moiety", "amine group" and simply "amine" are used
herein to describe a -NR'R" group or a -NR'- group, depending on its location
within
the molecule, where R' and R" are each independently hydrogen, alkyl,
cycloalkyl,
aryl, heteroaryl or heterocyclic, as these terms are defined hereinbelow.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
The polyamine chelator can be a linear polyamine, a cyclic polyamine or a
combination thereof.
A linear polyamine, according to the present invention, can be a polyamine
that has a general formula I:
5
HX-Am-(Y~B~)~ ~~~~(YnBn)n-ZH
Formula I
wherein m is an integer from 1 to 10; n is an integer from 0 to 20; X and Z
are
l0 each independently selected from the group consisting of an oxygen atom, a
sulfur
atom and a -NH group; Y, and Yn are each independently selected from the group
consisting of an oxygen atom, a sulfur atom and a -NH group; A is an alkylene
chain
having between 1 and 10 substituted and/or non-substituted carbon atoms; and
B, and
Bn are each independently an alkylene chain having between 1 and 20
substituted
15 and/or non-substituted carbon atoms, provided that at least one of X, Z, Y,
and Yn is
a -NH group and/or at least one of the carbon atoms in the alkylene chains is
substituted by an amine group.
Hence, the linear polyamine, according to the present invention, is preferably
comprised of one or more alkylene chains (Am, B,--~-Bn, in Formula I), is
interrupted
20 by one or more heteroatoms such as S, O and N (Y ~ ~ ~ ~ ~Yn in Formula I),
and
terminates with two such heteroatoms (X and Z in Formula I).
Alkylene chain A, as is described hereinabove, includes 1-10 substituted or
non-substituted carbon atoms and is connected, at least at one end thereof, to
a
heteroatom (e.g., X in Formula I). Whenever there are more than one alkylene
chains
25 A (in cases where m is greater than one), only the first alkylene chain A
is connected
to X. However, m is preferably 1 and hence the linear polyamine depicted in
Formula
I preferably includes only one alkylene chain A.
Alkylene chain B, as is described hereinabove, includes between 1 and 20
substituted or non-substituted carbon atoms. The alkylene chain B is connected
at its
30 two ends to a heteroatom (Y, ~ ~ ~ ~Yn and Z in Formula I).
The preferred linear polyamine delineated in Formula I comprises between 1
and 20 alkylene chains B, denoted as B, ~~-- Bn, where "B~ ~~~~ Bn" is used
herein to
describe a plurality of alkylene chains B, namely, B,, BZ, B3, ~~~~, Bn-1 and
Bn, where



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
56
n equals 0-20. These alkylene chains can be the same or different. Each of B,
~~~~ Bn
is connected to the respective heteroatom Y, ~~~~ Yn, and the last alkylene
chain in the
structure, Bn, is also connected to the heteroatom Z.
It should be noted that herein throughout, whenever an integer equals 0 or
whenever a component of a formula is followed by the digit 0, this component
is
absent from the structure. For example, if n in Formula I equals 0, there is
no
alkylene chain B and no heteroatom Y are meant.to be in the structure.
Preferably, n equals 2-10, more preferably 2-8 and most preferably 3-5.
Hence, the linear polyamine depicted in Formula I preferably includes between
3 and
5 alkylene chains B, each connected to 3-5 heteroatoms Y.
The linear polyamine depicted in Formula I must include at least one amine
group, as this term is defined hereinabove, preferably at least two amine
groups and
more preferably at least four amine groups. The amine group can be present in
the
structure as the heteroatoms X, Z or Y, ~~~~ Yn, such that at least one of X,
Z and Y,
~~~~ Yn is a -NH- group, or as a substituent of one or more of the substituted
carbon
atoms in the alkylene chains A and B, ~~~~ Bn. The presence of these amine
groups is
required in order to form a stable chelate with the copper ion, as is
discussed
hereinabove.
The alkylene chain A preferably has a general Formula II:
R, R2 Rg
C ~ g-CZH ....... C~H_
Formula II
wherein g is an integer that equals 0 or 3-10.
Hence, the alkylene chain A is comprised of a plurality of carbon atoms C,,
C2, C3 ~~~~, Cg-1 and Cg, substituted by the respective R,, R2, R3 ~~~~, Rg-1
and Rg
groups. Preferably, the alkylene chain A includes 2-10 carbon atoms, more
preferably, 2-6 and most preferably 2-4 carbon atoms.
As is defined hereinabove, in cases where g equals 0, the component CgH(Rg)
is absent from the structure and hence the alkylene chain A comprises only 2
carbon
atoms.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
57
R,, RZ and Rg are each a substituent attached to the carbon atoms in A. Each
of R,, RZ and Rg can independently be a substituent such as, but not limited
to,
hydrogen, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroalicyclic,
heteroaryl, halo,
amino, alkylamino, arylamino, cycloalkylamino, heteroalicyclic amino,
heteroarylamino, hydroxy, alkoxy, aryloxy, azo, C-amido, N-amido, ammonium,
thiohydroxy, thioalkoxy, thioaryloxy, sulfonyl, sulfinyl, N-sulfonamide, S-
sulfonamide, phosphonyl, phosphinyl, phosphonium, carbonyl, thiocarbonyl, C-
carboxy, O-carboxy, C-thiocarboxy, O-thiocarboxy, N-carbamate, O-carbamate, N-
thiocarbamate, O-thiocarbamate, urea, thiourea, borate, borane, boroaza,
silyl, siloxy,
silaza, aquo, alcohol, peroxo, amine oxide, hydrazine, alkyl hydrazine, aryl
hydrazine,
nitric oxide, cyanate, thiocyanate, isocyanate, isothiocyanate, cyano,
alkylnitrile, aryl
nitrile, alkyl isonitrile, aryl isonitrile, nitrate, nitrite, azido, alkyl
sulfonic acid, aryl
sulfonic acid, alkyl sulfoxide, aryl sulfoxide, alkyl aryl sulfoxide, alkyl
sulfenic acid,
aryl sulfenic acid, alkyl sulfinic acid, aryl sulfuric acid, alkyl thiol
carboxylic acid,
aryl thiol carboxylic acid, alkyl thiol thiocarboxylic acid, aryl thiol
thiocarboxylic
acid, carboxylic acid, alkyl carboxylic acid, aryl carboxylic acid, sulfate,
sulfite,
bisulfate, thiosulfate, thiosulfite, alkyl phosphine, aryl phosphine, alkyl
phosphine
oxide, aryl phosphine oxide, alkyl aryl phosphine oxide, alkyl phosphine
sulfide, aryl
phosphine sulfide, alkyl aryl phosphine sulfide, alkyl phosphonic acid, aryl
phosphonic acid, alkyl phosphinic acid, aryl phosphinic acid, phosphate,
thiophosphate, phosphate, pyrophosphate, triphosphate, hydrogen phosphate,
dihydrogen phosphate, guanidino, S-dithiocarbamate, N-dithiocarbamate,
bicarbonate,
carbonate, perchlorate, chlorate, chlorite, hypochlorite, perbromate, bromate,
bromite,
hypobromite, tetrahalomanganate, tetrafluoroborate, hexafluoroantimonate,
hypophosphite, iodate, periodate, metaborate, tetraarylborate, tetraalkyl
borate,
tartarate, salicylate, succinate, citrate, ascorbate, saccharirate, amino
acid, hydroxamic
acid and thiotosylate.
Whenever R,, Rz or Rg is hydrogen, its respective carbon atom in a non-
substituted carbon atom.
As used herein, the term "alkyl" is a saturated aliphatic hydrocarbon
including
straight chain and branched chain groups. Preferably, the alkyl group has 1 to
20
carbon atoms. More preferably, it is a medium size alkyl having 1 to 10 carbon
atoms. Most preferably, it is a lower alkyl having 1 to 4 carbon atoms. The
alkyl



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
58
group may be substituted or non-substituted. When substituted, the substituent
group
can be, for example, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy,
alkoxy,
aryloxy, thiohydroxy, thioalkoxy, thioaryloxy, cyano, halo, carbonyl,
thiocarbonyl, O-
carbamate, N-carbamate, O-thiocarbamate, N-thiocarbamate, C-amido, N-amido, C-
carboxy, O-carboxy, nitro, sulfonamide, silyl, guanidine, urea or amino, as
these
terms are defined hereinbelow.
The term "alkenyl" describes an alkyl group which consists of at least two
carbon atoms and at least one carbon-carbon double bond.
The term "alkynyl" describes an alkyl group which consists of at least two
carbon atoms and at least one carbon-carbon triple bond.
The term "cycloalkyl" describes an all-carbon monocyclic or fused ring (i.e.,
rings which share an adjacent pair of carbon atoms) group wherein one of more
of the
rings does not have a completely conjugated pi-electron system. Examples,
without
limitation, of cycloalkyl groups are cyclopropane, cyclobutane, cyclopentane,
cyclopentene, cyclohexane, cyclohexadiene, cycloheptane, cycloheptatriene, and
adamantane. A cycloalkyl group may be substituted or unsubstituted. When
substituted, the substituent group can be, for example, alkyl, aryl,
heteroaryl,
heteroalicyclic, hydroxy, alkoxy, aryloxy, thiohydroxy, thioalkoxy,
thioaryloxy,
cyano, halo, carbonyl, thiocarbonyl, C-carboxy, O-carboxy, O-carbamate, N-
2o carbamate, C-amido, N-amido, nitro, or amino, as these terms are defined
hereinabove
or hereinbelow.
The term "aryl" describes an all-carbon monocyclic or fused-ring polycyclic
(i.e., rings which share adjacent pairs of carbon atoms) groups having a
completely
conjugated pi-electron system. Examples, without limitation, of aryl groups
are
phenyl, naphthalenyl and anthracenyl. The aryl group may be substituted or
unsubstituted. When substituted, the substituent group can be, for example,
halo,
trihalomethyl, alkyl, hydroxy, alkoxy, aryloxy, thiohydroxy, thiocarbonyl, C-
carboxy,
O-carboxy, O-carbamate, N-carbamate, O-thiocarbamate, N-thiocarbamate, C-
amido,
N-amido, sulfinyl, sulfonyl or amino, as these terms are defined hereinabove
or
3o hereinbelow.
The term "heteroaryl" describes a monocyclic or fused ring (i.e., rings which
share an adjacent pair of atoms) group having in the rings) one or more atoms,
such
as, for example, nitrogen, oxygen and sulfur and, in addition, having a
completely



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
59
conjugated pi-electron system. Examples, without limitation, of heteroaryl
groups
include pyrrole, furane, thiophene, imidazole, oxazole, thiazole, pyrazole,
pyridine,
pyrimidine, quinoline, isoquinoline and purine. The heteroaryl group may be
substituted or unsubstituted. When substituted, the substituent group can be,
for
example, alkyl, cycloalkyl, halo, trihalomethyl, hydroxy, alkoxy, aryloxy,
thiohydroxy, thiocarbonyl, sulfonamide, C-carboxy, O-carboxy, sulfinyl,
sulfonyl, O-
carbamate, N-carbamate, O-thiocarbamate, N-thiocarbamate, C-amido, N-amido or
amino, as these terms are defined hereinabove or hereinbelow.
The term "heteroalicyclic" describes a monocyclic or fused ring group having
in the rings) one or more atoms such as nitrogen, oxygen and sulfur. The rings
may
also have one or more double bonds. However, the rings do not have a
completely
conjugated pi-electron system. The heteroalicyclic may be substituted or
unsubstituted. When substituted, the substituted group can be, for example,
alkyl,
cycloalkyl, aryl, heteroaryl, halo, trihalomethyl, hydroxy, alkoxy, aryloxy,
thiohydroxy, thioalkoxy, thioaryloxy, cyano, nitro, carbonyl, thiocarbonyl, C-
carboxy,
O-carboxy, O-carbamate, N-carbamate, O-thiocarbamate, N-thiocarbamate,
sulfinyl,
sulfonyl, C-amido, N-amido or amino, as these terms are defined hereinabove or
hereinbelow.
The term "halo" describes a fluorine, chlorine, bromine or iodine atom.
The term "amino", as is defined hereinabove with respect to an "amine" or an
"amino group", is used herein to describe an -NR'R", wherein R' and R" are
each
independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl or heterocyclic,
as these
terms are defined hereinabove.
Hence, the terms "alkylamino", "arylamino", "cycloalkylamino",
"heteroalicyclic amino" and "heteroarylamino" describe an amino group, as
defined
hereinabove, wherein at least one of R' and R" thereof is alkyl, aryl,
cycloalkyl,
heterocyclic and heteroaryl, respectively.
The term "hydroxy" describes an -OH group.
An "alkoxy" describes both an -O-alkyl and an -O-cycloalkyl group, as
3o defined herein.
An "aryloxy" describes both an -O-aryl and an -O-heteroaryl group, as defined
herein.
The term "azo" describes a -N=N group.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
A "C-amido" describes a -C(=O)-NR'R" group, where R' and R" are as
defined hereinabove.
An "N-amido" describes a R'C(=O)-NR"- group, where R' and R" are as
defined hereinabove.
5 An "ammonium" describes an -N+HR'R" group, where R' and R" are as
defined hereinabove.
The term "thiohydroxy" describes a -SH group.
The term "thioalkoxy" describes both a -S-alkyl group and a -S-cycloalkyl
group, as defined hereinabove.
10 The term "thioaryloxy" describes both a -S-aryl and a -S-heteroaryl group,
as
defined hereinabove.
A "sulfinyl" describes a -S(=O)-R group, where R can be, without limitation,
alkyl, cycloalkyl, aryl and heteroaryl as these terms are defined hereinabove.
A "sulfonyl" describes a -S(=O)2-R group, where R is as defined hereinabove.
15 A "S-sulfonamido" is a -S(=O)z-NR'R" group, with R' and R" as defined
hereinabove.
A "N-sulfonamido" is an R'(S=O)Z-NR"- group, with R' and R" as defined
hereinabove.
A "phosphonyl" is a -O-P(=O)(OR')-R" group, with R' and R" as defined
20 hereinabove.
A "phosphinyl" is a -PR'R" group, with R' and R" as defined hereinabove.
A "phosphonium" is a -P+R'R"R"', where R' and R" are as defined
hereinabove and R"' is defined as either R' or R".
The term "carbonyl" describes a -C(=O)-R group, where R is hydrogen, alkyl,
25 cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) or
heteroalicyclic (bonded
through a ring carbon) as defined hereinabove.
A "thiocarbonyl" describes a -C(=S)-R group, where R is as defined
hereinabove with respect to the term "carbonyl".
A "C-carboxy" describes a -C(=O)-O-R groups, where R is as defined
30 hereinabove with respect to the term "carbonyl".
An "O-carboxy" group refers to a RC(=O)-O- group, where R is as defined
hereinabove with respect to the term "carbonyl".
A "carboxylic acid" is a C-carboxy group in which R is hydrogen.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
61
A "C-thiocarboxy" is a -C(=S)-O-R groups, where R is as defined hereinabove
with respect to the term "carbonyl".
An "O-thiocarboxy" group refers to an R-C(=S)-O- group, where R is as
defined hereinabove with respect to the term "carbonyl".
The term "O-carbamate" describes an -OC(=O)-NR'R" group, with R' and
R" as defined hereinabove.
A "N-carbamate" describes a R'-O-C(=O)-NR"- group, with R' and R" as
defined hereinabove.
An "O-thiocarbamate" describes an -O-C(=S)-NR'R" group, with R' and R"
as defined hereinabove.
A "N-thiocarbamate" describes a R'OC(=S)NR"- group, with R' and R" as
defined hereinabove.
The term "urea" describes a -NR'-C(=O)-NR'R" group, with R', R" and R"'
as defined hereinabove.
The term "thiourea" describes a -NR'-C(=S)-NR'R" group, with R', R" and
R"' as defined hereinabove.
The term "borate" describes an -O-B-(OR)2 group, with R as defined
hereinabove.
The term "borane" describes a -B-R'R" group, with R' and R" as defined
hereinabove.
The term "boraza" describes a -B(R')(NR"R"') group, with R', R" and R"'
as defined hereinabove.
The term "silyl" describes a -SiR'R"R"', with R', R" and R"' as defined
herein.
The term "siloxy" is a -Si-(OR)3, with R as defined hereinabove.
The term "silaza" describes a -Si-(NR'R")3, with R' and R" as defined
herein.
The term "aquo" describes a HZO group.
The term "alcohol" describes a ROH group, with R as defined hereinabove.
3o The term "peroxo" describes an -OOR group, with R as defined hereinabove.
As used herein, an "amine oxide" is a -N(=O)R'R"R"' group, with R', R"
and R"' as defined herein.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
62
A "hydrazine" is a -NR'-NR"R"' group, with R', R" and R"' as defined
herein.
Hence, "alkyl hydrazine" and "aryl hydrazine" describe a hydrazine where R'
is an alkyl or an aryl, respectively, and R" and R"' are as defined
hereinabove.
The term "nitric oxide" is a -N=O group.
The term "cyano" is a -C---N group.
A "cyanate" is an -O-C---N group.
A "thiocyanate" is a "-S-C---N group.
An "isocyanate" is a -N=C=O group.
to An "isothiocyanate" is a -N=C=S group.
The terms "alkyl nitrite" and "aryl nitrite" describe a -R-C---N group, where
R
is an alkyl or an aryl, respectively.
The terms "alkyl isonitrile" and "aryl isonitrile" describe a R-N---C- group,
where R is an alkyl or aryl, respectively.
A "nitrate" or "nitro" is a -NOZ group.
A "nitrite" is an -O-N=O group.
An "azido" is a N3+ group.
An "alkyl sulfonic acid" and an "aryl sulfonic acid" describe a -R-SOZ-OH
group, with R being an alkyl or an aryl, respectively.
2o An "alkyl sulfoxide", an "aryl sulfoxide" and an "alkyl aryl sulfoxide"
describe a -R'S(=O)R" group, where R' and R" are each an alkyl, R' and R" are
each an aryl and where R' is and alkyl and R" is an aryl, respectively.
An "alkyl sulfenic acid" and "aryl sulfenic acid" describe a -R-S-OH group,
where R is an alkyl or an aryl, respectively.
An "alkyl sulfinic acid" and "aryl sulfinic acid" describe a -R-S(=O)-OH
group where R is an alkyl or an aryl, respectively.
As used herein, the terms "alkyl carboxylic acid" and "aryl carboxylic acid"
describe a -R-C(=O)-OH group, where R is an alkyl or an aryl, respectively.
An "alkyl thiol carboxylic acid" and an "aryl thiol carboxylic acid" describe
a
3o -R-C(=O)-SH group, where R is an alkyl or an aryl, respectively.
An "alkyl thiol thiocarboxylic acid" and an "aryl thiol thiocarboxylic acid"
describe a -R-C(=S)-SH group, where R is an alkyl or an aryl, respectively.
A "sulfate" is a -O-SOZ-OR' group, with R' as defined hereinabove.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
63
A "sulfite" group is a -O-S(=O)-OR' group, with R' as defined hereinabove.
A "bisulfate" is a sulfite group, where R' is hydrogen.
A "thiosulfate" is an -O-SOZ-SR' group, with R' as defined hereinabove.
A "thiosulfite" group is an -O-S(=O)-SR' group, with R' as defined
hereinabove.
The terms "alkyl/aryl phosphine" describe a -R-PHZ group, with R being an
alkyl or an aryl, respectively, as defined above.
The terms "alkyl and/or aryl phosphine oxide" describe a -R'-PR"Z(=O)
group, with R' and R" being an alkyl and/or an aryl, as defined hereinabove.
The terms "alkyl and/or aryl phosphine sulfide" describe a -R'-PR"2(=S)
group, with R' and R" being an alkyl and/or an aryl, as defined hereinabove.
The terms "alkyl/aryl phosphonic acid" describe a -R'-P(=O)(OH)2 group,
with R' being an alkyl or an aryl as defined above.
The terms "alkyl/aryl phosphinic acid" describes a -R'-P(OH)Z group, with R'
being an alkyl or an aryl as defined above.
A "phosphate" is a -O-P(=O)(OR')(OR") group, with R' and R" as defined
hereinabove.
A "hydrogen phosphate" is a phosphate group, where R' is hydrogen.
A "dihydrogen phosphate" is a phosphate group, where R' and R" are both
hydrogen.
A "thiophosphate" is a -S-P(=O)(OR')2 group, with R' as defined hereinabove.
A "phosphate" is an -O-P (OR')z group, with R' as defined hereinabove.
A "pyrophosphate" is an -O-P-(OR')-O-P(OR")Z group, with R' and R" as
defined hereinabove.
A "triphosphate" describes an -OP(=O)(OR')-O-P(=O)(OR")-O-
P(=O)(OR"')2, with R', R" and R"' are as defined hereinabove.
As used herein, the term "guanidine" describes a -R'NC(=N)-NR"R"' group,
with R', R" and R"' as def ned herein.
The term "S-dithiocarbamate" describes a -SC(=S)-NR'R" group, with R' and
R" as defined hereinabove.
The term "N-dithiocarbamate" describes an R' SC(=S)-NR"- group, with R'
and R" as defined hereinabove.
A "bicarbonate" is an -O-C(=O)-O- group.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
64
A "carbonate" is an -O-C(=O)-OH group.
A "perchlorate" is an -O-Cl(=O)3 group.
A "chlorate" is an -O-Cl(=O)2 group.
A "chlorite" is an -O-Cl(=O) group.
A "hypochlorite" is an -OCl group.
A "perbromate" is an -O-Br(=O)3 group.
A "bromate" is an -O-Br(=O)Z group..
A "bromite" is an -O-Br(=O) group.
A "hypobromite" is an -OBr group.
A "periodate" is an -O-I(=O)3 group.
A "iodate" is an -O-I(=O)Z group.
The term "tetrahalomanganate" describes MnCl4, MnBr4 and MnI4.
The term "tetrafluoroborate" describes a -BF4 group.
A "tetrafluoroantimonate" is a SbF~ group.
A "hypophosphite" is a -P(OH)2 group.
The term "metaborate" describes the group
R."O\B/O\8/OR,
I I
O~B~O
OR"
where R', R" and R"' are as defined hereinabove.
The terms "tetraalkyl/tetraaryl borate" describe a R'B- group, with R' being
an
alkyl or an aryl, respectively, as defined above.
A "tartarate" is an -OC(=O)-CH(OH)-CH(OH)-C(=O)OH group.
A "salycilate" is the group
coo-
~OH
A "succinate" is an -O-C(=O)-(CHZ)Z-COOH group.
A "citrate" is an -O-C(=O)-CHZ-CH(OH)(COOH)-CHZ-COOH group.
An "ascorbate" is the group
OH
I
-OH2CHC O O
HO \OH



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
A "saccharirate" is an oxidized saccharide having two carboxylic acid group.
The term "amino acid" as used herein includes natural and modified amino
acids and hence includes the 21 naturally occurring amino acids; those amino
acids
often modified post-translationally in vivo, including, for example,
hydroxyproline,
5 phosphoserine and phosphothreonine; and other unusual amino acids including,
but
not limited to, 2-aminoadipic acid, hydroxylysine, isodesmosine, nor-valine,
nor-
leucine and ornithine. Furthermore, the term "amino acid" includes both D- and
L-
amino acids which are linked via a peptide bond or a peptide bond analog to at
least
one addition amino acid as this term is defined herein.
10 A "hydroxamic acid" is a -C(=O)-NH-OH group.
A "thiotosylate" is the group
0
II
o=s-s-
I
CH3
Similarly, each of the alkylene chains B, ~~~~ Bn independently has a general
15 formula III:
Rp R(p+1) Rq
-Cp-C(p+1)H ww~CqH-
H
Formula III
2o wherein p is an integer that equals 0 or g+1 and q is an integer from g+2
to g+20.
Hence, each of the alkylene chains B, ~~~~ Bn is comprised of a plurality of
carbon atoms Cp, Cp+1, Cp+2 ~~-~, Cq-1 and Cq, substituted by the respective
Rp,
Rp+1, Rp+2 ~~~~, Rq-1 and Rq groups. Preferably, each of the alkylene chains
B, ~~~~
Bn includes 2-20 carbon atoms, more preferably 2-10, and most preferably 2-6
carbon
25 atoms.
As is defined hereinabove, in cases where p equals 0, the component -
CpH(Rp)- is absent from the structure. In cases where p equals g+1, it can be
either 1
or 4-11. The integer q can be either 2 or 5-20.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
66
Each of the substituents Rp, Rp+1 ~~~~ Rn can be any of the substituents
described hereinabove with respect to R,, RZ and Rg.
Hence, a preferred linear polyamine according to the present invention
includes two or more alkylene chains. The alkylene chains are interrupted
therebetween by a heteroatom and each is connected to a heteroatom at one end
thereof. Preferably, each of the alkylene chains include at least two carbon
atoms, so
as to enable the formation of a stable chelate between the heteroatoms and the
copper
ion.
The linear polyamine delineated in Formula I preferably includes at least one
to chiral carbon atom. Hence, at least one of C,, CZ and Cg in the alkylene
chain A
and/or at least one of Cp, Cp+1 and Cq in the alkylene chain B is chiral.
A preferred linear polyamine according to the present invention is
tetraethylenepentamine. Other representative examples of preferred linear
polyamines usable in the context of the present invention include, without
limitation,
ethylendiamine, diethylenetriamine, triethylenetetramine, triethylenediamine,
aminoethylethanolamine, pentaethylenehexamine, triethylenetetramine, N,N'-
bis(3-
aminopropyl)-1,3-propanediamine, and N,N'-Bis(2-animoethyl)-1,3
propanediamine.
In cases where the polyamine chelator is a cyclic polyamine, the polyamine
can have a general formula IV:
r-D
~X Am-(Y~B~)~---(YnBn)n-Z
Forinula IV
wherein m is an integer from 1 to 10; n is an integer from 0 to 20; X and Z
are each
independently selected from the group consisting of an oxygen atom, a sulfur
atom
and a -NH group; Y, and Yn are each independently selected from the group
consisting of an oxygen atom, a sulfur atom and a -NH group; A is an alkylene
chain
having between 1 and 10 substituted and/or non-substituted carbon atoms; B,
and Bn
are each independently an alkylene chain having between 1 and 20 substituted
and/or
non-substituted carbon atoms; and D is a bridging group having a general
formula V:



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
67
U-W-V
Formula V
whereas U and V are each independently selected from the group consisting of
substituted hydrocarbon chain and non-substituted hydrocarbon chain; and W is
selected from the group consisting of amide, ether, ester, disulfide,
thioether,
thioester, imine and alkene, provided that at least one of said X, Z, Y, and
Yn is a -
NH group and/or at least one of said carbon atoms in said alkylene chains is
substituted by an amine group.
l0 Optionally, the cyclic polyamine has one of the general formulas VI-X:
X IAm-(Y~Byi---(YnBn)n-ZH
Formula VI
D
H X-Am-(Y ~ B ~ ) i- - - (YnBn)n-Z
Formula VII
D---_T________1
X Am-(Y~B~)~---(YnBn)n-ZH
Formula VIII
r________
HX-Am-(Y ~'B ~ ) ~- - - (YnBn)n-Z
Formula IX
______
HX-Am-(Y 1 B 1 ) 1- - - (YnBn)n-ZH
Formula X



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
68
wherein m, n, X, Y,, Yn, Z, A, B and D are as described above and further
wherein
should the bridging group D is attached at one end to A (Formulas VI, VII and
X), U
or V are being attached to one carbon atom in the alkylene chain and should D
is
attached at one end to B 1 or Bn (Formulas VIII, IX and X), U or V are being
attached
to one carbon atom in the alkylene chain.
Hence, a preferred cyclic polyamine according to the present invention
includes two or more alkylene chains, A, B, ~~~~ Bn, as is detailed
hereinabove with
respect to the linear polyamine. The alkylene chains can form a cyclic
structure by
being connected, via the bridging group D, between the ends thereof, namely
between
the heteroatoms X and Z (Formula IV). Optionally, the alkylene chains can form
a
conformationally restricted cyclic structure by being connected, via the
bridging
group D, therebetween (Formula X). Further optionally, a conformationally
restricted
cyclic structure can be formed by connecting one alkylene chain to one
terminal
heteroatom (X or Z, Formulas VI-IX).
As is described hereinabove, in cases where the cyclic structure is formed by
connecting one alkylene chain to one terminal heteroatom, as is depicted in
Formulas
VI-IX, the bridging group D connects a terminal heteroatom, namely X or Z, and
one
carbon atom in the alkylene chains A and B, ~~~~ Bn. This carbon atom can be
anyone
of C,, C2, Cg, Cp, Cp+1 and Cq described hereinabove.
2o As is further described hereinabove, the cyclic structure is formed by the
bridging group D, which connects two components in the structure. The bridging
group D has a general formula U-W-V, where each of U and V is a substituted or
non-
substituted hydrocarbon chain.
As used herein, the phrase "hydrocarbon chain" describes a plurality of carbon
atoms which are covalently attached one to another and are substituted, inter
alia, by
hydrogen atoms. The hydrocarbon chain can be saturated, unsaturated, branched
or
unbranched and can therefore include one or more alkyl, alkenyl, alkynyl,
cycloalkyl
and aryl groups and combinations thereof.
The length of the hydrocarbon chains, namely the number of carbon atoms in
3o the chains, is preferably determined by the structure of the cyclic
polyamine, such that
on one hand, the ring tension of the formed cyclic structure would be
minimized and
on the other hand, an efficient chelation with the copper ion would be
achieved.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
69
When the hydrocarbon chain is substituted, the substituents can be any one or
combinations of the substituents described hereinabove with respect to R~, Rz
and Rg
in the linear polyamine.
The two hydrocarbon chains are connected therebetween by the group W,
which can be amide, ether, ester, disulfide, thioether, thioester, imine and
alkene.
As used herein, the term "ether" is an -O- group.
The term "ester" is a -C(=O)-O- group.
A "disulfide" is a -S-S- group.
A "thioether" is a -S- group.
A "thioester" is a -C(=O)-S- group.
An "imine" is a -C(=NH)- group.
An "alkene" is a -CH=CH- group.
The bridging group D is typically formed by connecting reactive derivatives of
the hydrocarbon chains U and V, so as to produce a bond therebetween (W), via
well-
known techniques, as is described, for example, in U.S. Patent No. 5,811,392.
As is described above with respect to the linear polyamine, the cyclic
polyamine must include at least one amine group, preferably at least two amine
groups and more preferably at least four amine groups, so as to form a stable
copper
chelate.
A preferred cyclic polyamine according to the present invention is cyclam
( 1,4,8,11-tetraazacyclotetradecane).
As is described hereinabove, the polyamine chelator of the present invention
can further include a multimeric combination of one or more linear
polyamine(s) and
one or more cyclic polyamine(s). Such a polyamine chelator can therefore be
comprised of any combinations of the linear and cyclic polyamines described
hereinabove.
Preferably, such a polyamine chelator has a general Formula XI:
~(Eyf LQ~-(Gyg~~n-f(Ez)i-LQ2-(G2)u~k_......._ f(EnO-LQ"-(Gn)o~~t
Formula XI
wherein n is an integer greater than 1; each of f, g, h, i, j, k, l, o and t
is independently
an integer from 0 to 10; each of E,, EZ and En is independently a linear
polyamine, as



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
is described hereinabove; each of G,, GZ and Gn is independently a cyclic
polyamine
as is described hereinabove; and each of Q,, QZ and Qn is independently a
linker
linking between two of said polyamines, provided that at least one of said Q,,
Qz and
Qn is an amine group and/or at least one of said linear polyamine and said
cyclic
5 polyamine has at least one free amine group.
Each of E,, EZ and En in Formula XI represent a linear polyamine as is
described in detail hereinabove, while each of G~, GZ and Gn represents a
cyclic
polyamine as is described in detail hereinabove.
The polyamine described in Formula XI can include one or more linear
l0 polyamine(s), each connected to another linear polyamine or to a cyclic
polyamine.
Each of the linear or cyclic polyamines in Formula XI is connected to another
polyamine via one or more linker(s), represented by Q,, QZ and Qn in Formula
XI.
Each of the linkers) Q,, QZ and Qn can be, for example, alkylene, alkenylene,
alkynylene, arylene, cycloalkylene, hetroarylene, amine, azo, amide, sulfonyl,
15 sulfinyl, sulfonamide, phosphonyl, phosphinyl, phosphonium, ketoester,
carbonyl,
thiocarbonyl, ester, ether, thioether, carbamate, thiocarbamate, urea,
thiourea, borate,
borane, boroaza, silyl, siloxy and silaza.
As used herein, the term "alkenylene" describes an alkyl group which consists
of at least two carbon atoms and at least one carbon-carbon double bond.
20 The term "alkynylene" describes an alkyl group which consists of at least
two
carbon atoms and at least one carbon-carbon triple bond.
The term "cycloalkylene" describes an all-carbon monocyclic or fused ring
(i.e., rings which share an adjacent pair of carbon atoms) group wherein one
of more
of the rings does not have a completely conjugated pi-electron system.
Examples,
25 without limitation, of cycloalkyl groups are cyclopropane, cyclobutane,
cyclopentane,
cyclopentene, cyclohexane, cyclohexadiene, cycloheptane, cycloheptatriene, and
adamantane.
The term "arylene" describes an all-carbon monocyclic or fused-ring
polycyclic (i.e., rings which share adjacent pairs of carbon atoms) groups
having a
3o completely conjugated pi-electron system. Examples, without limitation, of
aryl
groups are phenyl, naphthalenyl and anthracenyl. The aryl group may be
substituted
or unsubstituted.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
71
The term "heteroarylene" describes a monocyclic or fused ring (i.e., rings
which share an adjacent pair of atoms) group having in the rings) one or more
atoms,
such as, for example, nitrogen, oxygen and sulfur and, in addition, having a
completely conjugated pi-electron system. Examples, without limitation, of
heteroaryl groups include pyrrole, furane, thiophene, imidazole, oxazole,
thiazole,
pyrazole, pyridine, pyrimidine, quinoline, isoquinoline and purine. The
heteroaryl
group may be substituted or unsubstituted.
As used in the context of the linker of the present invention, the term
"amine"
describes an -NR'-, wherein R' can be hydrogen, alkyl, cycloalkyl, aryl,
heteroaryl or
heterocyclic, as these terms are defined hereinabove.
As is further used in the context of the linker of the present invention, the
term
"azo" describes a -N=N- group.
The term "amide" describes a -C(=O)-NR'- group, where R' is as defined
hereinabove.
The term "ammonium" describes an -N+HR'- group, where R' is as defined
hereinabove.
The term "sulfinyl" describes a -S(=O)- group.
The term "sulfonyl" describes a -S(=O)Z- group.
The term "sulfonamido" describes a -S(=O)Z-NR'- group, with R' as defined
hereinabove.
The term "phosphonyl" describes a -O-P(=O)(OR')- group, with R' as defined
hereinabove.
The term "phosphinyl" describes a -PR'- group, with R' as defined
hereinabove.
The term "phosphonium" is a -P+R'R", where R' and R" are as defined
hereinabove.
The term "ketoester" describes a -C(=O)-C(=O)-O- group.
The term "carbonyl" describes a -C(=O)- group.
The term "thiocarbonyl" describes a -C(=S)- group.
The term "carbamate" describes an -OC(=O)-NR'- group, with R' as defined
hereinabove.
The term "thiocarbamate" describes an -OC(=S)-NR- group, with R' as
defined hereinabove.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
72
The term "urea" describes an -NR'-C(=O)-NR"- group, with R' and R" and
as defined hereinabove.
The term "thiourea" describes a -NR'-C(=S)-NR'- group, with R' and R" as
defined hereinabove.
The term "borate" describes an -O-B-(OR)- group, with R as defined
hereinabove.
The term "borane" describes a -B-R-'- group, with R as defined hereinabove.
The term "boraza" describes a -B (NR'R")- group, with R' and R" as defined
hereinabove.
to The term "silyl" describes a -SiR'R"-, with R' and R" as defined herein.
The term "siloxy" is a -Si-(OR)Z-, with R as defined hereinabove.
The term "silaza" describes a -Si-(NR'R")Z-, with R' and R" as defined
herein.
It should be noted that all the terms described hereinabove in the context of
the
linker of the present invention are the same as described above with respect
to the
substituents. However, in distinction from the substituent groups, which are
connected to a component at one end thereof, the linker groups are connected
to two
components at two sites thereof and hence, these terms have been redefined
with
respect to the linker.
As has been mentioned hereinabove, according to the presently most preferred
embodiment of the present invention, the polyamine chelator is
tetraethylenepentamine (TEPA). However, other preferred polyamine chelators
include, without limitation, ethylendiamine, diethylenetriamine,
triethylenetetramine,
triethylenediamine, aminoethylethanolamine, aminoethylpiperazine,
pentaethylenehexamine, triethylenetetramine, captopril, penicilamine, N,N'-
bis(3-
aminopropyl)-1,3-propanediamine, N,N'-Bis(2-animoethyl)-1,3-propanediamine,
1,7-
dioxa-4,10-diazacyclododecane, 1,4,8,11-tetraazacyclotetradecane-5,7-dione,
1,4,7-
triazacyclononane, 1-oxa-4,7,10-triazacyclododecane, 1,4,8,12-
tetraazacyclopentadecane and 1,4,7,10-tetraazacyclododecane.
The above listed preferred chelators are known in their high affinity towards
copper ions. However, these chelators are further beneficially characterized
by their
substantial affinity also towards other transition metals, as is described by
Ross and
Frant [Ross JW and Frant MS. Chelometric indicators, titration with the solid-
state



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
73
cupric ion selective electrode. Analytical Chemistry 41:1900, 1969], which is
incorporated by reference as if fully set forth herein.
All the polyamine chelators described hereinabove can be either commercially
obtained or can be synthesized using known procedures such as described, for
example, in: T.W. Greene (ed.), 1999 ("Protective Groups in Organic Synthesis"
3ed
Edition, John Wiley & Sons, Inc., New York 779 pp); or in: R.C. Larock and
V.C.H.
Wioley, "Comprehensive Organic Transformations - A Guide to Functional Group
Preparations", (1999) 2°d Edition.
A preferred procedure for preparing tetraethylenepentamine-copper chelate
(TEPA-Cu) is described in PCT/IL03/00062.
The copper chelate or chelator can be provided to the cell culture medium.
The final concentrations of copper chelate may be, depending on the specific
application, in the micromolar or millimolar ranges, for example, within about
0.1 pM
to about 100 mM, preferably within about 4 ~M to about 50 mM, more preferably
within about S ~M to about 40 mM.
The methods described hereinabove for ex-vivo expanding hematopoietic stem
cell populations result, inter alia, in an expanded population of
hematopoietic stem
cells.
Thus, further according to an aspect of the present invention there are
provided
ex-vivo expanded populations of hematopoietic stem cells, obtained by any of
the
methods described hereinabove. The expanded populations of hematopoietic stem
cells according to the present invention comprise a plurality of cells
characterized by
3-20 % of the cells being reselectable CD34+ cells, of which at least 40 % of
cells are
CD34+~;",, i.e., fall below the median intensity in a FAGS analysis, wherein,
in the
reselectable CD34+ cells, a majority of cells which are Liri are also CD34+d;m
cells.
In one embodiment, the population of hematopoietic stem cells has a single
genetic background.
In another embodiment, the ex-vivo expanded population of hematopoietic
stem cells comprises at least N cells derived from a single donor, wherein N
equals
3o the average number of CD34+ cells derived from one sample of hematopoietic
mononuclear cells, multiplied by 1,000.
Cell surface expression of the CD34 and/or Lin markers can be determined,
for example, via FACS analysis or immunohistological staining techniques. A
self



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
74
renewal potential of the hematopoietic stem cells can be determined in-vitro
by long
term colony formation (LTC-CFUc), as is further exemplified in the Examples
section
that follows.
As is discussed in detail hereinabove, ex-vivo expansion of hematopoietic stem
cells can be advantageously utilized in various applications such as, for
example,
hematopoietic cells transplantation or implantation, adoptive immunotherapy
and
gene therapy. The ability to practice the ex-vivo expansion of hematopoietic
stem
cells with hematopoietic mononuclear cells as the cells source substantially
facilitates
the utilization of the methods described hereinabove in these applications.
Hence, according to another aspect of the present invention there is provided
a
method of hematopoietic cells transplantation or implantation. The method
according
to this aspect of the present invention is effected by (a) obtaining
hematopoietic
mononuclear cells which comprise a major fraction of hematopoietic committed
cells
and a minor fraction of hematopoietic stem and progenitor cells from a donor,
(b)
providing the hematopoietic mononuclear cells with ex-vivo culture conditions
for cell
proliferation and, at the same time, for reducing an expression and/or
activity of
CD38, so as to expand a population of the hematopoietic stem cells, while at
the same
time, substantially inhibiting differentiation of the hematopoietic stem cells
ex-vivo,
and (c) transplanting or implanting the thus obtained hematopoietic stem cells
to a
recipient.
As is described hereinabove, various agents can be used in the context of the
different aspects of the present invention for reducing an expression and/or
activity of
CD38.
Thus, in a particular embodiment of this aspect of the present invention, the
method is effected by providing the hematopoietic mononuclear cells with ex-
vivo
culture conditions for cell proliferation and, at the same time, for reducing
a capacity
of the hematopoietic mononuclear cells in responding to retinoic acid,
retinoids and/or
Vitamin D, as is described hereinabove.
In another particular embodiment, the method is effected by providing the
hematopoietic mononuclear cells with ex-vivo culture conditions for cell
proliferation
and, at the same time, for reducing a capacity of the hematopoietic
mononuclear cells
in responding to signaling pathways involving the retinoic acid receptor, the
retinoid
X receptor and/or the Vitamin D receptor, as is described hereinabove.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
In another particular embodiment of this aspect of the present invention, the
method is effected by providing the hematopoietic mononuclear cells with ex-
vivo
culture conditions for cell proliferation and, at the same time, for reducing
a capacity
of the hematopoietic mononuclear cells in responding to signaling pathways
involving
5 PI 3-kinase, as is described hereinabove.
In still another particular embodiment of this aspect of the present
invention,
the method is effected by providing the hematopoietic mononuclear cells with
ex-vivo
culture conditions for cell proliferation and with nicotinamide, a
nicotinamide analog,
a nicotinamide or a nicotinamide analog derivative or a nicotinamide or a
to nicotinamide analog metabolite, as is described hereinabove.
In another particular embodiment of this aspect of the present invention, the
method is effected by providing the hematopoietic mononuclear cells with ex-
vivo
culture conditions for cell proliferation and with a PI 3-kinase inhibitor, as
is
described hereinabove.
15 In another aspect of the present invention, the method of hematopoietic
cells
transplantation or implantation described above is effected by providing the
hematopoietic mononuclear cells with ex-vivo culture conditions for cell
proliferation
and with one or more of the copper chelator(s) or chelate(s) described
hereinabove.
In any of the methods of this aspect of the present invention, the donor and
the
20 recipient can be a single individual or different individuals, for example,
allogeneic or
xenogeneic individuals. When allogeneic transplantation is practiced, regimes
for
reducing implant rejection and/or graft vs. host disease, as well know in the
art,
should be undertaken. Such regimes are currently practiced in human therapy.
Most
advanced regimes are disclosed in publications by Slavin S. et al., e.g., J
Clin
25 Immunol (2002) 22: 64, and J Hematother Stem Cell Res (2002) 11: 265), Gur
H. et
al. (Blood (2002) 99: 4174), and Martelli MF et al, (Semin Hematol (2002) 39:
48),
which are incorporated herein by reference.
The methods described hereinabove can be utilized to produce transplantable
hematopoietic cell preparations, such that according to yet another aspect of
the
3o present invention there is provided a transplantable hematopoietic cell
preparation,
which comprises an expanded population of hematopoietic stem cells propagated
ex-
vivo from hematopoietic mononuclear cells which comprise, prior to expansion,
a
major fraction of hematopoietic committed cells and a minor fraction of



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
76
hematopoietic stem and progenitor cells, in the presence of an effective
amount of an
agent for reducing the expression and/or activity of CD38, while at the same
time,
substantially inhibiting differentiation of said hematopoietic stem cells, and
a
pharmaceutically acceptable carrier.
As is described hereinabove, various agents were found to reduce the
expression and/or activity of CD38, while at the same time, substantially
inhibit
differentiation of the hematopoietic stem cells under these conditions.
Hence, in a particular embodiment of this aspect of the present invention, the
agent described above is an agent that reduces a capacity of the hematopoietic
mononuclear cells in responding to retinoic acid, retinoids and/or Vitamin D,
while at
the same time, substantially inhibits differentiation of the hematopoietic
stem cells.
In another particular embodiment of this aspect of the present invention, the
agent described above is an agent that reduces a 'capacity of the
hematopoietic
mononuclear cells in responding to retinoic acid receptor, retinoid X receptor
and/or
Vitamin D receptor signaling, while at the same time, substantially inhibits
differentiation of the stem cells.
In yet another particular embodiment of this aspect of the present invention,
the agent described above is an agent that reduces a capacity of the
hematopoietic
mononuclear cells in responding to PI 3-kinase signaling, while at the same
time,
substantially inhibits differentiation of the stem cells.
In still another particular embodiment of this aspect of the present
invention,
the agent described above comprises an effective amount of an agent selected
from
the group consisting of nicotinamide, a nicotinamide analog, a nicotinamide or
a
nicotinamide analog derivative and a nicotinamide or a nicotinamide analog
metabolite.
In still another particular embodiment of this aspect of the present
invention,
the agent described above comprises an effective amount of a PI 3-kinase
inhibitor.
According to still another aspect of the present invention there is provided a
transplantable hematopoietic cell preparation, which comprises an expanded
population of hematopoietic stem cells propagated ex-vivo from hematopoietic
mononuclear cells which comprise, prior to expansion, a major fraction of
hematopoietic committed cells and a minor fraction of hematopoietic stem and
progenitor cells, in the presence of at least one copper chelate or chelator,
as defined



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
77
hereinabove, while at the same time, substantially inhibiting differentiation
of said
hematopoietic stem cells, and a pharmaceutically acceptable carrier.
As is further discussed hereinabove, the ex-vivo expansion of hematopoietic
stem cells of the present invention can be utilized in adoptive immunotherapy.
Similarly to the hematopoietic transplantation or implementation methods of
the present invention, a method of adoptive therapy according to the present
invention
is effected by (a) obtaining hematopoietic mononuclear cells which comprise a
major
fraction of hematopoietic committed cells and a minor fraction of
hematopoietic stem
and progenitor cells from a recipient; (b) providing the hematopoietic
mononuclear
cells with ex-vivo culture conditions for cell proliferation and, at the same
time, with
each of the copper chelators or chelates described hereinabove and/or each of
the
agents for reducing the expression and/or activity of CD38 described
hereinabove, so
as to expand the population of the hematopoietic stem cells, while at the same
time,
substantially inhibiting differentiation of the hematopoietic stem cells, as
is detailed
hereinabove; and (c) transplanting the thus obtained hematopoietic stem cells
to the
recipient.
As is further detailed below, stem cells in general and hematopoietic stem
cells
in particular may serve to exert cellular gene therapy.
Gene therapy as used herein refers to the transfer of genetic material (e.g.,
2o DNA or RNA) of interest into a host to treat or prevent a genetic or
acquired disease
or condition or phenotype. The genetic material of interest encodes a product
(e.g., a
protein, polypeptide, peptide, functional RNA, antisense) whose production in
vivo is
desired. For example, the genetic material of interest can encode a hormone,
receptor,
enzyme, polypeptide or peptide of therapeutic value. For review see, in
general, the
text "Gene Therapy" (Advanced in Pharmacology 40, Academic Press, 1997).
Two basic approaches to gene therapy have evolved: (i) ex-vivo or cellular
gene therapy; and (ii) in vivo gene therapy. In ex-vivo gene therapy cells are
removed
from a patient, and while being cultured are treated in-vitro. Generally, a
functional
replacement gene is introduced into the cells via an appropriate gene delivery
3o vehicle/method (transfection, transduction, homologous recombination, etc.)
and an
expression system as needed and then the modified cells are expanded in
culture and
returned to the host/patient. These genetically re-implanted cells have been
shown to
express the transfected genetic material in situ.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
78
Hence, further according to an aspect of the present invention, there is
provided a method of genetically modifying stem cells with an exogene. The
method,
according to this aspect of the present invention, is effected by (a)
obtaining
hematopoietic mononuclear cells which comprise a major fraction of
hematopoietic
committed cells and a minor fraction of hematopoietic stem and progenitor
cells, (b)
providing the hematopoietic mononuclear cells with ex-vivo culture conditions
for cell
proliferation and, at the same time, with each of the copper chelators or
chelates
described hereinabove and/or each of the agents for reducing the expression
and/or
activity of CD38 described hereinabove, so as to expand the population of the
to hematopoietic stem cells, while at the same time, substantially inhibiting
differentiation of the hematopoietic stem cells, as is detailed hereinabove,
and (c)
genetically modifying the hematopoietic stem cells with the exogene.
In a preferred embodiment, genetically modifying the cells is effected by a
vector, which comprises the exogene or transgene, which vector is, for
example, a
viral vector or a nucleic acid vector. Many viral vectors suitable for use in
cellular
gene therapy are known, examples are provided hereinbelow. Similarly, a range
of
nucleic acid vectors can be used to genetically transform the expanded cells
of the
invention, as is further described below.
Accordingly, the expanded cells of the present invention can be modified to
express a gene product. As used herein, the phrase "gene product" refers to
proteins,
peptides and functional RNA molecules. Generally, the gene product encoded by
the
nucleic acid molecule is the desired gene product to be supplied to a subject.
Examples of such gene products include proteins, peptides, glycoproteins and
lipoproteins normally produced by an organ of the recipient subject. For
example,
gene products which may be supplied by way of gene replacement to defective
organs
in the pancreas include insulin, amylase, protease, lipase, trypsinogen,
chymotrypsinogen, carboxypeptidase, ribonuclease, deoxyribonuclease,
triaclyglycerol lipase, phospholipase A2, elastase, and amylase; gene products
normally produced by the liver include blood clotting factors such as blood
clotting
Factor VIII and Factor IX, UDP glucuronyl transferae, ornithine
transcarbanoylase,
and cytochrome p450 enzymes, and adenosine deaminase, for the processing of
serum
adenosine or the endocytosis of low density lipoproteins; gene products
produced by
the thymus include serum thymic factor, thymic humoral factor, thymopoietin,
and



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
79
thymosin al; gene products produced by the digestive tract cells include
gastrin,
secretin, cholecystokinin, somatostatin, serotinin, and substance P.
Alternatively, the encoded gene product is one, which induces the expression
of the desired gene product by the cell (e.g., the introduced genetic material
encodes a
transcription factor, which induces the transcription of the gene product to
be supplied
to the subject).
In still another embodiment, the recombinant gene can provide a heterologous
protein, e.g., not native to the cell in which it is expressed. For instance,
various
human MHC components can be provided to non-human cells to support engraftment
1o in a human recipient. Alternatively, the transgene is one, which inhibits
the
expression or action of a donor MHC gene product.
A nucleic acid molecule introduced into a cell is in a form suitable for
expression in the cell of the gene product encoded by the nucleic acid.
Accordingly,
the nucleic acid molecule includes coding and regulatory sequences required
for
transcription of a gene (or portion thereof) and, when the gene product is a
protein or
peptide, translation of the gene acid molecule include promoters, enhancers
and
polyadenylation signals, as well as sequences necessary for transport of an
encoded
protein or peptide, for example N-terminal signal sequences for transport of
proteins
or peptides to the surface of the cell or secretion.
Nucleotide sequences which regulate expression of a gene product (e.g.,
promoter and enhancer sequences) are selected based upon the type of cell in
which
the gene product is to be expressed and the desired level of expression of the
gene
product. For example, a promoter known to confer cell-type specific expression
of a
gene linked to the promoter can be used. A promoter specific for myoblast gene
expression can be linked to a gene of interest to confer muscle-specific
expression of
that gene product. Muscle-specific regulatory elements, which are known in the
art,
include upstream regions from the dystrophin gene (Klamut et al., (1989) Mol.
Cell
Biol.9: 2396), the creatine kinase gene (Buskin and Hauschka, (1989) Mol. Cell
Biol.
9: 2627) and the troponin gene (Mar and Ordahl, (1988) Proc. Natl. Acad. Sci.
USA.
85: 6404). Regulatory elements specific for other cell types are known in the
art (e.g.,
the albumin enhancer for liver-specific expression; insulin regulatory
elements for
pancreatic islet cell-specific expression; various neural cell-specific
regulatory
elements, including neural dystrophin, neural enolase and A4 amyloid
promoters).



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
Alternatively, a regulatory element, which can direct constitutive expression
of
a gene in a variety of different cell types, such as a viral regulatory
element, can be
used. Examples of viral promoters commonly used to drive gene expression
include
those derived from polyoma virus, Adenovirus 2, cytomegalovirus and Simian
Virus
5 40, and retroviral LTRs.
Alternatively, a regulatory element, which provides inducible expression of a
gene linked thereto, can be used. The use of an inducible regulatory element
(e.g., an
inducible promoter) allows for modulation of the production of the gene
product in
the cell. Examples of potentially useful inducible regulatory systems for use
in
10 eukaryotic cells include hormone-regulated elements (e.g., see Mader, S.
and White,
J.H. (1993) Proc. Natl. Acad. Sci. USA 90: 5603-5607), synthetic ligand-
regulated
elements (see, e.g., Spencer, D.M. et al. 1993) Science 262: 1019-1024) and
ionizing
radiation-regulated elements (e.g., see Manome, Y. Et al. (1993)
Biocherrcistry 32:
10607-10613; Datta, R. et al. (1992) Proc. Natl. Acad. Sci. USA 89: 1014-
10153).
~ 5 Additional tissue-specific or inducible regulatory systems, which may be
developed,
can also be used in accordance with the invention.
There are a number of techniques known in the art for introducing genetic
material into a cell that can be applied to modify a cell of the invention.
In one embodiment, the nucleic acid is in the form of a naked nucleic acid
2o molecule. In this situation, the nucleic acid molecule introduced into a
cell to be
modified consists only of the nucleic acid encoding the gene product and the
necessary regulatory elements.
Alternatively, the nucleic acid encoding the gene product (including the
necessary regulatory elements) is contained within a plasmid vector. Examples
of
25 plasmid expression vectors include CDM8 (Seed, B. (1987) Nature 329: 840)
and
pMT2PC (Kaufman, et al. (1987) EMBOJ. 6: 187-195).
In another embodiment, the nucleic acid molecule to be introduced into a cell
is contained within a viral vector. In this situation, the nucleic acid
encoding the gene
product is inserted into the viral genome (or partial viral genome). The
regulatory
3o elements directing the expression of the gene product can be included with
the nucleic
acid inserted into the viral genome (i.e., linked to the gene inserted into
the viral
genome) or can be provided by the viral genome itself.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
81
Naked nucleic acids can be introduced into cells using calcium phosphate
mediated transfection, DEAF-dextran mediated transfection, electroporation,
liposome-mediated transfection, direct injection, and receptor-mediated
uptake.
Naked nucleic acid, e.g., DNA, can be introduced into cells by forming a
precipitate containing the nucleic acid and calcium phosphate. For example, a
HEPES-buffered saline solution can be mixed with a solution containing calcium
chloride and nucleic acid to form a precipitate and the precipitate is then
incubated
with cells. A glycerol or dimethyl sulfoxide shock step can be added to
increase the
amount of nucleic acid taken up by certain cells. CaP04-mediated transfection
can be
used to stably (or transiently) transfect cells and is only applicable to in
vitro
modification of cells. Protocols for CaP04-mediated transfection can be found
in
Current Protocols in Molecular Biology, Ausubel, F.M. et al. (eds.) Greene
Publishing
Associates, (1989), Section 9.1 and in Molecular Cloning: A Laboratory Manual,
2nd
Edition, Sambrook et al. Cold Spring Harbor Laboratory Press, (1989), Sections
16.32-16.40 or other standard laboratory manuals.
Naked nucleic acid can be introduced into cells by forming a mixture of the
nucleic acid and DEAF-dextran and incubating the mixture with the cells. A
dimethylsulfoxide or chloroquine shock step can be added to increase the
amount of
nucleic acid uptake. DEAE-dextran transfection is only applicable to in vitro
modification of cells and can be used to introduce DNA transiently into cells
but is
not preferred for creating stably transfected cells. Thus, this method can be
used for
short-term production of a gene product but is not a method of choice for long-
term
production of a gene product. Protocols for DEAF-dextran-mediated transfection
can
be found in Current Protocols in Molecular Biology, Ausubel, F.M. et al.
(eds.)
Greene Publishing Associates (1989), Section 9.2 and in Molecular Cloning: A
Laboratory Manual, 2nd Edition, Sambrook et al. Cold Spring Harbor Laboratory
Press, (1989), Sections 16.41-16.46 or other standard laboratory manuals.
Naked nucleic acid can also be introduced into cells by incubating the cells
and the nucleic acid together in an appropriate buffer and subjecting the
cells to a
high-voltage electric pulse. The efficiency with which nucleic acid is
introduced into
cells by electroporation is influenced by the strength of the applied field,
the length of
the electric pulse, the temperature, the conformation and concentration of the
DNA
and the ionic composition of the media. Electroporation can be used to stably
(or



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
82
transiently) transfect a wide variety of cell types and is only applicable to
in vitro
modification of cells. Protocols for electroporating cells can be found in
Current
Protocols in Molecular Biology, Ausubel F.M. et al. (eds.) Greene Publishing
Associates, ( 1989), Section 9.3 and in Molecular Cloning: A Laboratory
Manual, 2nd
Edition, Sambrook et al. Cold Spring Harbor Laboratory Press, ( 1989),
Sections
16.54-16.55 or other standard laboratory manuals.
Another method by which naked nucleic acid can be introduced into cells
includes liposome-mediated transfection (lipofection). The nucleic acid is
mixed with
a liposome suspension containing cationic lipids. The DNA/liposome complex is
then
incubated with cells. Liposome mediated transfection can be used to stably (or
transiently) transfect cells in culture in vitro. Protocols can be found in
Current
Protocols in Molecular Biology, Ausubel F.M. et al. (eds.) Greene Publishing
Associates, (1989), Section 9.4 and other standard laboratory manuals.
Additionally,
gene delivery in vivo has been accomplished using liposomes. See for example
Nicolau et al. (1987) Meth. Enz. 149:157-176; Wang and Huang (1987) Proc.
Natl.
Acad. Sci. USA 84:7851-7855; Brigham et al. (1989) Am. J Med. Sci. 298:278;
and
Gould-Fogerite et al. (1989) Gene 84:429-438.
Naked nucleic acid can also be introduced into cells by directly injecting the
nucleic acid into the cells. For an in vitro culture of cells, DNA can be
introduced by
2o microinjection. Since each cell is microinjected individually, this
approach is very
labor intensive when modifying large numbers of cells. However, a situation
wherein
microinjection is a method of choice is in the production of transgenic
animals
(discussed in greater detail below). In this situation, the DNA is stably
introduced into
a fertilized oocyte, which is then allowed to develop into an animal. The
resultant
animal contains cells carrying the DNA introduced into the oocyte. Direct
injection
has also been used to introduce naked DNA into cells in vivo (see e.g., Acsadi
et al.
(1991) Nature 332:815-818; Wolff et al. (1990) Science 247:1465-1468). A
delivery
apparatus (e.g., a "gene gun") for injecting DNA into cells in vivo can be
used. Such
an apparatus is commercially available (e.g., from BioRad).
3o Naked nucleic acid can be complexed to a canon, such as polylysine, which
is
coupled to a ligand for a cell-surface receptor to be taken up by receptor-
mediated
endocytosis (see for example Wu, G. and Wu, C.H. (1988) J. Biol. Chem. 263:
14621;
Wilson et al. (1992) J. Biol. Chem. 267: 963-967; and U.S. Patent No.
5,166,320).



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
83
Binding of the nucleic acid-ligand complex to the receptor facilitates uptake
of the
DNA by receptor-mediated endocytosis. Receptors to which a DNA-ligand complex
has targeted include the transferrin receptor and the asialoglycoprotein
receptor. A
DNA-ligand complex linked to adenovirus capsids which naturally disrupt
endosomes, thereby releasing material into the cytoplasm can be used to avoid
degradation of the complex by intracellular lysosomes (see for example Curiel
et al.
(1991) Proc. Natl. Acad. Sci. USA 88: 8850; Cristiano et al. (1993) Proc.
Natl. Acad.
Sci. USA 90: 2122-2126). Receptor-mediated DNA uptake can be used to introduce
DNA into cells either in vitro or in vivo and, additionally, has the added
feature that
1o DNA can be selectively targeted to a particular cell type by use of a
ligand which
binds to a receptor selectively expressed on a target cell of interest.
Generally, when naked DNA is introduced into cells in culture (e.g., by one of
the transfection techniques described above) only a small fraction of cells
(about 1 out
of 105) typically integrate the transfected DNA into their genomes (i.e., the
DNA is
maintained in the cell episomally). Thus, in order to identify cells, which
have taken
up exogenous DNA, it is advantageous to transfect nucleic acid encoding a
selectable
marker into the cell along with the nucleic acids) of interest. Preferred
selectable
markers include those, which confer resistance to drugs such as 6418,
hygromycin
and methotrexate. Selectable markers may be introduced on the same plasmid as
the
2o genes) of interest or may be introduced on a separate plasmid.
A preferred approach for introducing nucleic acid encoding a gene product
into a cell is by use of a viral vector containing nucleic acid, e.g., a cDNA,
encoding
the gene product. Infection of cells with a viral vector has the advantage
that a large
proportion of cells receive the nucleic acid which can obviate the need for
selection of
cells which have received the nucleic acid. Additionally, molecules encoded
within
the viral vector, e.g., a cDNA contained in the viral vector, are expressed
efficiently in
cells which have taken up viral vector nucleic acid and viral vector systems
can be
used either in vitro or in vivo.
Defective retroviruses are well characterized for use in gene transfer for
gene
therapy purposes (for review see Miller, A.D. (1990) Blood 76: 271). A
recombinant
retrovirus can be constructed having a nucleic acid encoding a gene product of
interest inserted into the retroviral genome. Additionally, portions of the
retroviral
genome can be removed to render the retrovirus replication defective. The
replication



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
84
defective retrovirus is then packaged into virions, which can be used to
infect a target
cell through the use of a helper virus by standard techniques. Protocols for
producing
recombinant retroviruses and for infecting cells in vitro or in vivo with such
viruses
can be found in Current Protocols in Molecular Biology, Ausubel, F.M. et al.
(eds.)
Greene Publishing Associates, (1989), Sections 9.10-9.14 and other standard
laboratory manuals. Examples of suitable retroviruses include pLJ, pZIP, pWE
and
pEM, which are well known to those skilled in the art. Examples of suitable
packaging virus lines include yrCrip, yrCrip, yr2 and yrAm. Retroviruses have
been
used to introduce a variety of genes into many different cell types, including
epithelial
1o cells endothelial cells, lymphocytes, myoblasts, hepatocytes, bone marrow
cells, in
vitro and/or in vivo (see for example Eglitis, et al. (1985) Science 230: 1395-
1398;
Danosand Mulligan (1988) Proc. Natl. Acad. Sci. USA 85: 6460-6464; Wilson et
al.
( 1988) Proc. Natl. Acad. Sci USA 85:3014-3018; Armentano et al., ( 1990)
Proc. Natl.
Acad. Sci. USA 87: 6141-6145; Huber et al. (1991) Proc. Natl. Acad. Sci. USA
88:
8039-8043; Feri et al. (1991) Proc. Natl. Acad. Sci. USA 88:8377-8381;
Chowdhury
et al. (1991) Science 254: 1802-1805; van Beusechem et al. (1992) Proc. Natl.
Acad.
Sci USA 89:7640-7644; Kay et al. (1992) Human Gene Therapy 3:641-647; Dai et
al.
(1992) Proc. Natl. Acad. Sci. USA 89:10892-10895; Hwu et al. (1993) J.
Immunol.
150:4104-4115; US Patent No. 4,868,116; US Patent No. 4,980,286; PCT
Application
WO 89/07136; PCT Application WO 89/02468; PCT Application WO 89/05345; and
PCT Application WO 92/07573). Retroviral vectors require target cell division
in
order for the retroviral genome (and foreign nucleic acid inserted into it) to
be
integrated into the host genome to stably introduce nucleic acid into the
cell. Thus, it
may be necessary to stimulate replication of the target cell.
The genome of an adenovirus can be manipulated such that it encodes and
expresses a gene product of interest but is inactivated in terms of its
ability to
replicate in a normal lytic viral life cycle. See for example Berkner et al.
(1988)
BioTechniques 6:616; Rosenfeld et al. (1991) Science 252:431-434; and
Rosenfeld et
al. (1992) Cell 68:143-155. Suitable adenoviral vectors derived from the
adenovirus
strain Ad type 5 d1324 or other strains of adenovirus (e.g., Ad2, Ad3, Ad7
etc.) are
well known to those skilled in the art. Recombinant adenoviruses are
advantageous in
that they do not require dividing cells to be effective gene delivery vehicles
and can
be used to infect a wide variety of cell types, including airway epithelium
(Rosenfeld



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
et al. (1992) cited supra), endothelial cells (Lemarchand et al. (1992) Proc.
Natl.
Acad. Sci. USA 89: 6482-6486), hepatocytes (Herz and Gerard (1993) Proc. Natl.
Acad. Sci. USA 90: 2812-2816) and muscle cells (Quantin et al. (1992) Proc.
Natl.
Acad. Sci. USA 89: 2581-2584). Additionally, introduced adenoviral DNA (and
5 foreign DNA contained therein) is not integrated into the genome of a host
cell but
remains episomal, thereby avoiding potential problems that can occur as a
result of
insertional mutagenesis in situations where introduced DNA becomes integrated
into
the host genome (e.g., retroviral DNA). Moreover, the carrying capacity of the
adenoviral genome for foreign DNA is large (up to 8 kilobases) relative to
other gene
to delivery vectors (Berkner et al. cited supra; Haj-Ahmand and Graham (1986)
J. Virol
57: 267). Most replication-defective adenoviral vectors currently in use are
deleted for
all or parts of the viral E 1 and E3 genes but retain as much as 80% of the
adenoviral
genetic material.
Adeno-associated virus (AAV) is a naturally occurring defective virus that
15 requires another virus, such as an adenovirus or a herpes virus, as a
helper virus for
efficient replication and a productive life cycle. (For a review see Muzyczka
et al.
Curr. Topics In Micro. And Immunol. (1992) 158: 97-129). It is also one of the
few
viruses that may integrate its DNA into non-dividing cells, and exhibits a
high
frequency of stable integration (see for example Flotte et al. (1992) Am. J.
Respir.
20 Cell. Mol. Biol. 7: 349-356; Samulski et al. (1989) J. Virol. 63:3822-3828;
and
McLaughlin et al. (1989) J. Virol. 62: 1963-1973). Vectors containing as
little as 300
base pairs of AAV can be packaged and can integrate. Space for exogenous DNA
is
limited to about 4.5 kb. An AAV vector such as that described in Tratschin et
al.
(1985) Mol. Cell. Biol. 5: 3251-3260 can be used to introduce DNA into cells.
A
25 variety of nucleic acids have been introduced into different cell types
using AAV
vectors (see for example Hermonat et al. ( 1984) Proc. Natl. Acad. Sci. USA
81: 6466-
6470; Tratschin et al. (1985) Mol. Cell Biol. 4: 2072-2081; Wondisford et al.
(1988)
Mol. Endocrinol. 2:32-39; Tratschin et al. (1984) J. Virol. S1: 611-619; and
Flotte et
al. (1993) J. Biol. Chem. 268: 3781-3790).
30 The efficacy of a particular expression vector system and method of
introducing nucleic acid into a cell can be assessed by standard approaches
routinely
used in the art. For example, DNA introduced into a cell can be detected by a
filter
hybridization technique (e.g., Southern blotting) and RNA produced by
transcription



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
86
of introduced DNA can be detected, for example, by Northern blotting, RNase
protection or reverse transcriptase-polymerase chain reaction (RT-PCR). The
gene
product can be detected by an appropriate assay, for example by immunological
detection of a produced protein, such as with a specific antibody, or by a
functional
assay to detect a functional activity of the gene product, such as an
enzymatic assay.
If the gene product of interest to be interest to be expressed by a cell is
not readily
assayable, an expression system can first be optimized using a reporter gene
linked to
the regulatory elements and vector to be used. The reporter gene encodes a
gene
product, which is easily detectable and, thus, can be used to evaluate
efficacy of the
l0 system. Standard reporter genes used in the art include genes encoding (3-
galactosidase, chloramphenicol acetyl transferase, luciferase and human growth
hormone.
When the method used to introduce nucleic acid into a population of cells
results in modification of a large proportion of the cells and efficient
expression of the
gene product by the cells (e.g., as is often the case when using a viral
expression
vector), the modified population of cells may be used without further
isolation or
subcloning of individual cells within the population. That is, there may be
sufficient
production of the gene product by the population of cells such that no further
cell
isolation is needed. Alternatively, it may be desirable to grow a homogenous
2o population of identically modified cells from a single modified cell to
isolate cells,
which efficiently express the gene product. Such a population of uniform cells
can be
prepared by isolating a single modified cell by limiting dilution cloning
followed by
expanding the single cell in culture into a clonal population of cells by
standard
techniques.
According to a preferred embodiment of the present invention, in each of the
methods described hereinabove, providing the hematopoietic mononuclear cells
with
conditions for ex-vivo cell proliferation is effected by providing the cells
with
nutrients and with cytokines. Preferably, the cytokines are early acting
cytokines,
such as, but not limited to, stem cell factor, FLT3 ligand, interleukin-1,
interleukin-2,
3o interleukin-3, interleukin-6, interleukin-10, interleukin-12, tumor
necrosis factor-a
and thrombopoietin. It will be appreciated in this respect that novel
cytokines are
continuously discovered, some of which may find uses in the methods of cell
expansion of the present invention.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
87
Late acting cytokines can also be used. These include, for example,
granulocyte colony stimulating factor, granulocyte/macrophage colony
stimulating
factor, erythropoietin, FGF, EGF, NGF, VEGF, LIF, Hepatocyte growth factor and
macrophage colony stimulating factor.
The ability of the agents of the present invention to inhibit differentiation
of
hematopoietic stem cells present in hematopoietic mononuclear cells can be
further
used in technical applications such as cells collection and cells culturing.
According to a further aspect of the present invention there is provided a
hematopoietic stem cells collection/culturing bag. The cells
collection/culturing bag
to of the present invention is supplemented with an effective amount of a
retinoic acid
receptor antagonist, a retinoid X receptor antagonist and/or a Vitamin D
receptor
antagonist, which substantially inhibits cell differentiation of a
hematopoietic stem
cells fraction of hematopoietic mononuclear cells which comprise a major
fraction of
hematopoietic committed cells and a minor fraction of hematopoietic stem and
progenitor cells. Alternatively, the hematopoietic stem cells
collection/culturing bag
of the present invention is supplemented with an effective amount of
nicotinamide, a
nicotinamide analog, a nicotinamide or a nicotinamide analog derivative or a
nicotinamide or a nicotinamide analog metabolite. Still alternatively, the
hematopoietic stem cells collection/culturing bag of the present invention is
supplemented with an effective amount of a PI 3-kinase inhibitor. Further
alternatively, the hematopoietic stem cells collection/culturing bag of the
present
invention is supplemented with an effective amount of one or more copper
chelator(s)
or chelate(s).
According to an additional aspect of the present invention, there is provided
an
assay of determining whether a specific molecule/agent, e.g., a retinoic acid
receptor
antagonist, a retinoid X receptor antagonist, a Vitamin D receptor antagonist,
a CD38
inhibitor, a PI 3-kinase inhibitor, a copper chelator or a copper chelate, is
an effective
agent for expanding a population of hematopoietic stem cells that are present
in a
hematopoietic mononuclear cells fraction.
The assay, according to this aspect of the present invention, is performed by
culturing hematopoietic mononuclear cells which comprise a major fraction of
hematopoietic committed cells and a minor fraction of hematopoietic stem and
progenitor cells in the presence of the tested agent/molecule and monitoring



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
88
expansion of the hematopoietic stem cells over time, e.g., a few weeks to a
few
months. If increased expansion and decreased differentiation occurs, as
compared to
non-treated cells, the tested agent/molecule is an effective hematopoietic
stem cell
expansion agent.
Preferably, culturing the hematopoietic mononuclear cells is performed in a
presence of an effective amount of a cytokine, preferably, an early acting
cytokine or
a combination of such cytokines, e.g., thrombopoietin (TPO), interleukin-6 (IL-
6), an
FLT-3 ligand and stem cell factor (SCF). This assay can be used, by one
ordinarily
skilled in the art, to determine, for example, which of the antagonists,
inhibitors or
copper chelators and chelates listed above is most efficient for the purpose
of
implementing the various methods and preparations of the present invention
described
hereinabove. The assay can be further used to determine most effective
concentrations and exposure time for achieving optimal results with
hematopoietic
mononuclear cells of different origins.
In each of the aspects of the present invention described hereinabove, the
hematopoietic mononuclear cells can be obtained from any multicellular
organism
including both animals and plants. Preferably, the hematopoietic mononuclear
cells
are obtained from the bone marrow (Rowley SD et al. (1998) Bone Marrow
Transplant 21: 1253), the peripheral blood (Koizumi K, (2000) Bone Marrow
Transplant 26: 787, the liver (Petersen BE et al. (1998) Hepatology 27: 433)
and
neonatal umbilical cord blood.
Additional objects, advantages, and novel features of the present invention
will
become apparent to one ordinarily skilled in the art upon examination of the
following
examples, which are not intended to be limiting. Additionally, each of the
various
embodiments and aspects of the present invention as delineated hereinabove and
as
claimed in the claims section below finds experimental support in the
following
examples.
3o EXAMPLES
Reference is now made to the following examples, which together with the
above descriptions, illustrate the invention in a non limiting fashion.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
89
EXAMPLE 1
THE EFFECT OF A COPPER CHELATOR ON THE EX VIVO EXPANSION
OF HEMATOPOIETIC STEM CELLS OF A MONONUCLEAR CELLS
CUL TURE
s
Experimental Procedures
Sample collection and processizzg: Samples were obtained from umbilical
cord blood after a normal full-term delivery and were frozen within 24 hours
pospartum. The blood cells were thawed in Dextran buffer and incubated for 15
to hours in MEM (Biological Industries, Israel) supplemented with 10 % fetal
calf serum
(FCS; Biological Industries). The cells were then layered on Ficoll-Hypaque
(density
1.077 gram/ml; Sigma) and centrifuged at 400 g for 30 minutes at room
temperature.
The mononuclear cells in the interface layer were then collected, washed three
times
in phosphate-buffered saline (PBS; Biological Industries), and re-suspended in
PBS
is containing 0.5 % human serum albumin (HSA). The cells were then split into
two
fractions, the first being the mononuclear cells (MNC) fraction and the second
fraction was used for purifying CD34+ cells by immunomagnetic separation using
the
"MiniMACS CD34+ progenitor cell isolation kit" (Miltenyi Biotec, Aubum, CA)
according to the manufacturer's recommendations. The purity of the CD34+ cells
20 obtained ranged between 95 % and 98 %, based on Flow Cytometry evaluation.
Ex-vivo expansion of lzematopoietic stem cells: The Mononuclear cells
(MNC), obtained as described hereinabove, were plated in 24-well Costar Cell
Clusters (Corning Inc., Corning, NY) or seeded in Culture Bags (American
Fluoroseal
Corp.), with alpha minimal essential medium (a-MEM) supplemented with 10 %
fetal
2s bovine serum (FBS, Biological Industries), at a concentration of about 10~
cells/ml.
The purified CD34+ cells were similarly plated or seeded in the Culture Bags,
at a
concentration of about 104 cellslml. The media were supplemented with
tetraethylpantamine (TEPA) chelator (obtained from Sigma) and/or with the
following human recombinant cytokines (all obtained from Perpo Tech, Inc.,
Rocky
30 Hill, NJ): Thrombopoietin (TPO), 50 ng/ml; interleukin 6 (IL-6), 50 ng/ml;
FLT-3
ligand, SO ng/ml and a stem cell factor (SCF), 50 ng/ml; occasionally SCF was
replaced by IL-3, 20 ng/ml. All cultures were incubated at 37 °C in an
atmosphere of



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
5 % C02 in air with extra humidity. At weekly intervals, the cell cultures
were semi-
depopulated and supplemented with fresh medium containing cytokines. Following
different incubation periods, cells were harvested, stained with trypan blue
and
enumerated.
5 Morphological assessment: Morphological characterization of the resulting
culture populations was accomplished on aliquots of cells deposited on glass
slides
via cytospin (Cytocentrifuge, Shandon, Runcorn, UK). Cells were fixed, stained
with
May-Grunwald/Giemsa stain and examined microscopically.
Surface antigen analysis: Cells were harvested, washed with a PBS solution
10 containing 1 % bovine sera albumin (BSA) and 0.1 % sodium azide (Sigma),
and
stained at 4 °C for 60 minutes with fluorescein isothiocyanate or
phycoerythrin-
conjugated antibodies (all from Immunoquality Products, the Netherlands). The
cells
were then washed with the same buffer and analyzed by FACS caliber or
Facstarplus
flow cytometers. Cells were passed at a rate of 1000 cells/second, using
saline as the
~ 5 sheath fluid. A 488 nm argon laser beam served as the light source for
excitation.
Emission of ten thousand cells was measured using logarithmic amplification,
and
analyzed using CellQuest software.
Determizzation of CD34+ cells and subsets: CD34+ surface expression on
short and long-term cultures initiated either with purified CD34+ cells or the
entire
20 MNC fraction was determined as follows: CD34+ cells were positively
reselected
(Miltenyi kit) and counted. Purity was confirmed by subsequent FACS and cell
morphology analysis, as is described hereinabove.
Reselected CD34+ cell subsets were stained for the following combination of
antigens: CD34PE/CD38FITC and CD34PE/38-, 33-, 14-, 15-, 3, 4, 61, 19 (Lin)
25 FITC.
Cell population calculations:
FACS analysis results are given as percentage values of cells. Absolute
numbers of subsets are calculated from the absolute number of CD34+cells.
Determination of baseline levels of CD34+/CD38- and CD34+/Liri cells was
30 conducted as follows: CD34+ cells were purified from 3 thawed cord blood
units and
stained for the above markers. The mean of these experiments was considered as
the
baseline value.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
91
Total cell counts, numbers of CD34+ cells and subsets, and CFU numbers are
presented as cumulative numbers, with the assumption that the cultures had not
been
passaged; i.e., the number of cells per ml were multiplied by the number of
passages
performed.
Assaying Colony Forming Unit (CFU) ability: Cells were cloned in semi-
solid, methylcellulose-containing medium supplemented with 2 IU/ml
erythropoietin
(Eprex, Cilag AG Int., Switzerland), stem cell factor and IL-3, both at 20
ng/ml, and
G-CSF and GM-CSF, both at 10 ng/ml (all from Perpo Tech). Cultures were
incubated for 14 days at 37 °C, 5 % C02 in a humidified atmosphere.
Determination of LTGCFUc values: The ability of the cultures to maintain
self renewal was measured by determination of the content of colony forming
unit
cells in the long and extended long-term cultures (LTC-CFUc), as described in
the
references hereinabove.
Experimental Results
Mononuclear cells (MNC) were seeded in culture bags and were provided
with nutrients and cytokines (50 ng/ml FLt3, IL-6, TPO and SCF) as described
above.
The MNC cultures were either treated or untreated (untreated controls) with
various
concentrations (S-10 pM) of TEPA chelator. The treated MNC cultures were
supplemented with TEPA for only the first three weeks and from week three
onward
were topped with chelator-free media. The pre-purified CD34+ cultures were not
supplemented with TEPA and served as positive controls. The cultures were
analyzed
weekly during a 12-week period for the number of cells, CFUc, CD34+ and
CD34+CD38- cells. In order to precisely determine the CD34+ cell content,
CD34+
cells were weekly reselected and enumerated from each of the experimental
groups
(treated and untreated MNC cultures) and the positive control (CD34+
cultures).
The results, illustrated in Figures 1 a-b, 2 and 3, show that addition of TEPA
chelator to non-purified MNC cultures, substantially and progressively
increased the
number of CD34+ cells, CD34+ colony-forming cells and CD34+CD38- cells, over a
12-week period. Thus, in MNC cultures treated with TEPA, the cumulative number
of CD34+ cells increased from a non-detectable level to over 8 x 10' cells/ml,
after 2
and 12 weeks, respectively (Figures la-b); the cumulative number of CD34+CD3g-



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
92
cells increased from a non-detectable level to 2.5 x 10~ cells/ml, after 2 and
12 weeks,
respectively (Figure 2); and the number of CD34+ CFUs increased from a non-
detectable level to 3.2 x 10' cells/ml after 2 and 10 weeks, respectively
(Figure 3).
On the other hand, when TEPA was not added to MNC cultures (untreated
controls),
no significant expansion of stem or progenitor cells was measured throughout
the 12-
week period. Furthermore, the stem and progenitor cells densities in the TEPA-
treated MNC cultures, either equalized or surpassed the densities of stem and
progenitor cells in pre-purified CD34+ cell cultures (not treated with TEPA,
positive
controls). Morphological analysis of cells derived from long-term and TEPA-
treated
MNC cultures, revealed a high proportion of non-differentiated cells, while
most of
the cells derived from long-term and MNC cultures not treated with TEPA, where
fully differentiated.
The results described in this Example clearly show that stem and progenitor
hematopoietic cells may be substantially expanded ex-vivo, continuously over
at least
12 weeks period, in a culture of mixed (mononuclear fraction) blood cells,
with no
prior purification of CD34+ cells. The data also show that this effect
resulted from
supplementing the cells culture medium with TEPA chelator, only during the
first
three weeks of culturing.
These results indicate that short-term MNC cell cultures supplemented with
2o TEPA in addition to cytokines, enabled tremendous expansion of CD34+ cells
and
stem/early progenitor cells (CD34+38-) as compared with minimal expansion of
these
cells obtained in MNC cultures treated only with cytokines. Comparison
experiments
demonstrated that expansion of CD34+ cells and its rare CD34+CD38- cell subset
continue to occur in the extended long-term cultures and is much higher as
compared
with that obtained from cultures initiated with highly purified CD34+ cells.
Therefore, the results may suggest that short-term treatment of MNC with TEPA
potentiate the MNC cultures in a way that enables higher expansion of cells
with
extended self renewal potential.
The results may also suggest that in addition to the regulatory effect on
3o CD34+ cells and its early subsets, the chelator may also enable ex-vivo
expansion of a
small subset of cells that are not co-purified with the CD34+ cell fraction.
This subset



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
93
of cells, which is probably in nature CD34-, may support superior expansion of
CD34+ cells and its subsets during the extended long-term cultures.
Hence, this Example illustrates a substantial ex-vivo expansion of stem and
progenitor cells in a mixed mononuclear cells culture. This novel procedure
circumvents the need of the laborious and costly enrichment of stem cells
prior to
initiation of cultures, which is currently used in the art. Hence, the use of
a copper
chelator, such as TEPA, can substantially simplify, reduce cost and improve
efficiency of procedures for an ex-vivo expansion of stem and/or progenitor
cells.
1 o EXAMPLE 2
THE EFFECT OF A COPPER CHELATE ON THE EX VIVO EXPANSION OF
HEMATOPOIETIC STEM CELLS OF A MONONUCLEAR CELLS CULTURE
Copper-TEPA chelate was prepared as described, for example, in
PCT/IL03/00062.
Mononuclear cells (MNC) were seeded in culture bags and were provided
with nutrients and cytokines as described in Example 1 above. The mononuclear
cell
cultures were either untreated (control) or treated with Cu-TEPA chelate. The
treated
MNC cultures were supplemented with Copper-TEPA chelate for the first three
weeks
and from week three onward were topped with chelator-free media. All cultures
were
2o analyzed eight weeks after an 8-week period.
The results, presented in Table 1 below, show that addition of Copper-TEPA
chelate to MNC cultures markedly increased the number of CD34+ cells, the
proportion of CD34+ cells, and the number of CD34+CD38- cells, after an eight
weeks incubation period. Thus, the cumulative number of CD34+ cells per
culture
bag after incubation was 2.56 x 106, 12.37 x 106 or 32.85 x 106, in the
untreated
cultures (cytokines only), 50 pM Copper-TEPA-treated and 100 ~M Copper-TEPA-
treated cultures, respectively. The cumulative number of CD34+CD3g- cells
increased from 2.1 x 105 in the untreated control culture (cytokines only) to
6.1 x 105
in the Copper-TEPA (100 pM) treated culture. ,



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
94
Table 1
Treatment Number of Portion of Number of
CD34+ cells CD34+ cells CD34/38- cells
(X 10 4) (%) (X 10 4)
Control 256.0 0.2 21
Cu-TEPA chelate 1237.3 1.4
SOpM
Cu-TEPA chelate 3285.3 1.2 61
100~M
The results described in this Example demonstrate that hematopoietic stem
cells may be substantially expanded ex-vivo, over at least 8 weeks period, in
a culture
of mononuclear blood cells, with no prior purification of CD34+ cells, in the
presence
of a copper chelate such as Copper-TEPA.
EXAMPLE 3
THE EFFECT OF A RAR ANTAGONIST ON THE EX VIVO EXPANSION OF
HEMATOPOIETIC STEM CELLS OF A MONONUCLEAR CELLS CULTURE
Materials and Experimental Methods
The high-Affinity retinoic acid receptor (RAR) antagonist 4-[[4-(4-
ethylphenyl)-2,2-dimethyl- (2H)-thiochomen-6-yl)]-benzoic acid, (AGN 194310)
was
synthesized according to the procedure described in PCT/IL03/00064.
Mononuclear cells fraction was collected and purified as described above in
Example 1. MNC cultures were prepared and maintained as described above. AGN
194310 RAR antagonist was added to the tested cultures at concentrations
ranging
from 1 x 10-3 - 1 x 10-~ ~ M [or 410 pg/1 to 4.1 x 10-5 pg/1]. The antagonist
was added
for a predetermined, limited period, for up to three weeks or continuously
during the
entire culture period.
The results, presented in Table 2, show that mononuclear cell fractions
cultured in the presence of RAR antagonists and cytokines revealed a
significant
increase in the number of CD34+Lin- cells (78 %, 24 %) as quantitated by FACS
analysis from a reselected, highly purified CD34+ cell fraction, as compared
with the



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
control untreated MNC fractions, 2 and 5 weeks (respectively), after initial
seeding.
The MNC cells responded to the RAR antagonists and expanded an
undifferentiated
population, without prior purification of the CD34+ population. RAR antagonist
treatment was sufficient to stimulate specific expansion of the
stem/progenitor cell
5 compartment, at 5 weeks post seeding. While control untreated MNCs had no
detectable CD34+ population, RAR antagonist treated cultures revealed
significant
numbers of CD34+ cells, and those that were lineage marker deficient. Thus,
any
factors elaborated by the MNC culture cells that suppress CD34+ cell survival
in
control samples are insufficient to override the signal provided by the RAR
antagonist
10 to elaborate this compartment.
Table 2
2 weeks


CytokinesCytokines +RAR
only antagonist 10-6
M


Ns of CD34 cells X 176 169
10


Ns of CD34+/Liri X 1.76 132.5
104


CD34/Liri 1 78.4


5 weeks


Cytokines Cytokines +RAR


only antagonist 10-6
M


N~ of CD34 cells 0 985
X 10'' *


N~ of CD34+/Lin- 0 237.8
X 10 *


CD34/Lin- 0 ~ 24.1 -_


It is appreciated that certain features of the invention, which are, for
clarity,
described in the context of separate embodiments, may also be provided in
combination in a single embodiment. Conversely, various features of the
invention,
which are, for brevity, described in the context of a single embodiment, may
also be
provided separately or in any suitable subcombination.



CA 02495824 2005-02-16
WO 2004/016731 PCT/IL2003/000681
96
Although the invention has been described in conjunction with specific
embodiments thereof, it is evident that many alternatives, modifications and
variations
will be apparent to those skilled in the art. Accordingly, it is intended to
embrace all
such alternatives, modifications and variations that fall within the spirit
and broad
scope of the appended claims. All publications, patents and patent
applications
mentioned in this specification are herein incorporated in their entirety by
reference
into the specification, to the same extent as if each individual publication,
patent or
patent application was specifically and individually indicated to be
incorporated herein
by reference. In addition, citation or identification of any reference in this
application
l0 shall not be construed as an admission that such reference is available as
prior art to
the present invention. The scope of the present invention and of the appended
claims is
not to be regarded as restricted or limited by or to any explicit or specific
theory
presented herein.

Representative Drawing

Sorry, the representative drawing for patent document number 2495824 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2003-08-17
(87) PCT Publication Date 2004-02-26
(85) National Entry 2005-02-16
Examination Requested 2008-06-02
Dead Application 2011-06-01

Abandonment History

Abandonment Date Reason Reinstatement Date
2010-06-01 R30(2) - Failure to Respond
2010-08-17 FAILURE TO PAY APPLICATION MAINTENANCE FEE

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2005-02-16
Maintenance Fee - Application - New Act 2 2005-08-17 $100.00 2005-02-16
Registration of a document - section 124 $100.00 2005-06-23
Maintenance Fee - Application - New Act 3 2006-08-17 $100.00 2006-05-19
Maintenance Fee - Application - New Act 4 2007-08-17 $100.00 2007-07-17
Maintenance Fee - Application - New Act 5 2008-08-18 $200.00 2008-05-27
Request for Examination $800.00 2008-06-02
Maintenance Fee - Application - New Act 6 2009-08-17 $200.00 2009-06-22
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
GAMIDA-CELL LTD.
Past Owners on Record
PELED, TONY
ROSEN, OREN
TREVES, AVI
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Abstract 2005-02-16 1 59
Claims 2005-02-16 62 2,691
Drawings 2005-02-16 2 37
Description 2005-02-16 96 5,000
Cover Page 2005-04-25 1 31
PCT 2005-02-16 7 361
Assignment 2005-02-16 4 147
Correspondence 2005-04-20 1 27
Assignment 2005-06-23 6 259
Prosecution-Amendment 2008-06-02 2 66
Prosecution-Amendment 2009-12-01 4 142